US20050208617A1 - N-acetylglucosamintransferase III expression in lower eukaryotes - Google Patents

N-acetylglucosamintransferase III expression in lower eukaryotes Download PDF

Info

Publication number
US20050208617A1
US20050208617A1 US10/680,963 US68096303A US2005208617A1 US 20050208617 A1 US20050208617 A1 US 20050208617A1 US 68096303 A US68096303 A US 68096303A US 2005208617 A1 US2005208617 A1 US 2005208617A1
Authority
US
United States
Prior art keywords
glcnac
pichia
host cell
man
activity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/680,963
Other versions
US7598055B2 (en
Inventor
Piotr Bobrowicz
Stephen Hamilton
Tilman Gerngross
Stefan Wildt
Byung-Kwon Choi
Juergen Nett
Robert Davidson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Glycofi Inc
Original Assignee
Piotr Bobrowicz
Hamilton Stephen R
Gerngross Tilman U
Stefan Wildt
Byung-Kwon Choi
Nett Juergen H
Davidson Robert C
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=32911943&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20050208617(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US09/892,591 external-priority patent/US7029872B2/en
Priority claimed from PCT/US2002/041510 external-priority patent/WO2003056914A1/en
Priority claimed from US10/371,877 external-priority patent/US7449308B2/en
Application filed by Piotr Bobrowicz, Hamilton Stephen R, Gerngross Tilman U, Stefan Wildt, Byung-Kwon Choi, Nett Juergen H, Davidson Robert C filed Critical Piotr Bobrowicz
Priority to US10/680,963 priority Critical patent/US7598055B2/en
Priority to PCT/US2004/005128 priority patent/WO2004074458A2/en
Priority to JP2006503757A priority patent/JP4611280B2/en
Priority to SI200431362T priority patent/SI1597381T2/en
Priority to AT04713412T priority patent/ATE449862T1/en
Priority to EP04713388.9A priority patent/EP1597381B2/en
Priority to CA2516550A priority patent/CA2516550C/en
Priority to AT04713388T priority patent/ATE452201T1/en
Priority to AU2004213868A priority patent/AU2004213868B2/en
Priority to US10/546,101 priority patent/US8697394B2/en
Priority to CA2753408A priority patent/CA2753408A1/en
Priority to DE602004024649T priority patent/DE602004024649D1/en
Priority to CA2516520A priority patent/CA2516520C/en
Priority to AT09004760T priority patent/ATE543912T1/en
Priority to JP2006503776A priority patent/JP4758335B2/en
Priority to SI200431310T priority patent/SI1599595T2/en
Priority to PCT/US2004/005191 priority patent/WO2004074461A2/en
Priority to ES04713388T priority patent/ES2336790T5/en
Priority to ES04713412T priority patent/ES2335345T5/en
Priority to PT04713388T priority patent/PT1597381E/en
Priority to EP04713412.7A priority patent/EP1599595B2/en
Priority to AU2004213859A priority patent/AU2004213859B2/en
Priority to EP09004760A priority patent/EP2080809B1/en
Priority to PT04713412T priority patent/PT1599595E/en
Priority to ES09004760T priority patent/ES2384929T3/en
Priority to DE602004024289T priority patent/DE602004024289D1/en
Publication of US20050208617A1 publication Critical patent/US20050208617A1/en
Assigned to GLYCOFI, INC. reassignment GLYCOFI, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GERNGROSS, TILLMAN U., NETT, JUERGEN HERMANN, BOBROWICZ, PIOTR, CHOI, BYUNG- KWON, DAVIDSON, ROBERT C., HAMILTON, STEPHEN R., WILDT, STEFAN
Priority to US12/313,636 priority patent/US20090226959A1/en
Priority to US12/540,915 priority patent/US20100016555A1/en
Priority to US12/540,849 priority patent/US8445227B2/en
Publication of US7598055B2 publication Critical patent/US7598055B2/en
Application granted granted Critical
Priority to AU2010201036A priority patent/AU2010201036B2/en
Priority to JP2010104525A priority patent/JP5255017B2/en
Priority to JP2010181319A priority patent/JP2011019521A/en
Priority to US14/340,621 priority patent/US20150079633A1/en
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/37Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi
    • C07K14/39Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi from yeasts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1048Glycosyltransferases (2.4)
    • C12N9/1051Hexosyltransferases (2.4.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/05Fusion polypeptide containing a localisation/targetting motif containing a GOLGI retention signal
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention is directed to methods and compositions by which non-human eukaryotic host cells, such as fungi or other eukaryotic cells, can be genetically modified to produce glycosylated proteins (glycoproteins) having patterns of glycosylation similar to those of glycoproteins produced by animal cells, especially human cells, which are useful as human or animal therapeutic agents.
  • non-human eukaryotic host cells such as fungi or other eukaryotic cells
  • glycosylation After DNA is transcribed and translated into a protein, further post-translational processing involves the attachment of sugar residues, a process known as glycosylation.
  • Different organisms produce different glycosylation enzymes (glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available, so that the glycosylation patterns as well as composition of the individual oligosaccharides, even of the same protein, will be different depending on the host system in which the particular protein is being expressed.
  • Bacteria typically do not glycosylate proteins, and if so only in a very unspecific manner (Moens and Vanderleyden (1997) Arch Microbiol. 168(3):169-175).
  • Lower eukaryotes such as filamentous fungi and yeast add primarily mannose and mannosylphosphate sugars.
  • the resulting glycan is known as a “high-mannose” type glycan or a mannan.
  • Plant cells and insect cells (such as Sf9 cells) glycosylate proteins in yet another way.
  • the nascent oligosaccharide side chain may be trimmed to remove several mannose residues and elongated with additional sugar residues that typically are not found in the N-glycans of lower eukaryotes. See, e.g., Bretthauer, et al.
  • Synthesis of a mammalian-type oligosaccharide structure begins with a set of sequential reactions in the course of which sugar residues are added and removed while the protein moves along the secretory pathway in the host organism.
  • the enzymes which reside along the glycosylation pathway of the host organism or cell determine the resulting glycosylation patterns of secreted proteins.
  • the resulting glycosylation pattern of proteins expressed in lower eukaryotic host cells differs substantially from the glycosylation pattern of proteins expressed in higher eukaryotes such as humans and other mammals (Bretthauer, 1999).
  • the structure of a typical fungal N-glycan is shown in FIG. 1A .
  • the early steps of human glycosylation can be divided into at least two different phases: (i) lipid-linked Glc 3 Man 9 GlcNAc 2 oligosaccharides are assembled by a sequential set of reactions at the membrane of the endoplasmic reticulum (ER) ( FIG. 13 ) and (ii) the transfer of this oligosaccharide from the lipid anchor dolichyl pyrophosphate onto de novo synthesized protein.
  • the site of the specific transfer is defined by an asparagine (Asn) residue in the sequence Asn-Xaa-Ser/Thr (SEQ ID NOs:1 and 2) where Xaa can be any amino acid except proline (Gavel and von Heijne (1990) Protein Eng.
  • glucosidases and mannosidases occurs in the ER before the nascent glycoprotein is transferred to the early Golgi apparatus, where additional mannose residues are removed by Golgi specific alpha ( ⁇ )-1,2-mannosidases. Processing continues as the protein proceeds through the Golgi.
  • a number of modifying enzymes including N-acetylglucosaminyl transferases (GnTI, GnTII, GnTIII, GnTIV and GnTV), mannosidase II and fucosyltransferases, add and remove specific sugar residues.
  • galactosyltranferases and sialyltransferases (ST) produce a glycoprotein structure that is released from the Golgi. It is this structure, characterized by bi-, tri- and tetra-antennary structures, containing galactose, fucose, N-acetylglucosamine and a high degree of terminal sialic acid, that gives glycoproteins their human characteristics.
  • the structure of a typical human N-glycan is shown in FIG. 1B . See also FIGS. 14 and 15 for steps involved in mammalian-type N-glycan processing.
  • glycoproteins are derived from a common lipid-linked oligosaccharide precursor Glc 3 Man 9 GlcNAc 2 -dolichol-pyrophosphate.
  • Glc 3 Man 9 GlcNAc 2 -dolichol-pyrophosphate Within the endoplasmic reticulum, synthesis and processing of dolichol pyrophosphate bound oligosaccharides are identical between all known eukaryotes.
  • further processing of the core oligosaccharide by fungal cells, e.g., yeast once it has been transferred to a peptide leaving the ER and entering the Golgi, differs significantly from humans as it moves along the secretory pathway and involves the addition of several mannose sugars.
  • the N-glycans of animal glycoproteins typically include galactose, fucose, and terminal sialic acid. These sugars are not found on glycoproteins produced in yeast and filamentous fungi.
  • nucleotide sugar precursors e.g., UDP-N-acetylglucosamine, UDP-N-acetylgalactosamine, CMP-N-acetylneuraminic acid, UDP-galactose, GDP-fucose, etc.
  • Glycosyl transfer reactions typically yield a side product which is a nucleoside diphosphate or monophosphate. While monophosphates can be directly exported in exchange for nucleoside triphosphate sugars by an antiport mechanism, diphosphonucleosides (e.g., GDP) have to be cleaved by phosphatases (e.g. GDPase) to yield nucleoside monophosphates and inorganic phosphate prior to being exported.
  • phosphatases e.g. GDPase
  • This reaction is important for efficient glycosylation; for example, GDPase from Saccharomyces cerevisiae ( S. cerevisiae ) has been found to be necessary for mannosylation. However that GDPase has 90% reduced activity toward UDP (Berninsone et al.
  • UDP is known to be a potent inhibitor of glycosyltransferases and the removal of this glycosylation side product may be important to prevent glycosyl-transferase inhibition in the lumen of the Golgi (Khatara et al. (1974) Eur. J. Biochem. 44:537-560). See Berninsone et al. (1995) J. Biol. Chem. 270(24):14564-14567; Beaudet et al. (1998) Abc Transporters: Biochemical, Cellular, and Molecular Aspects 292: 397-413.
  • Sugar transferases and glycosidases line the inner (luminal) surface of the ER and Golgi apparatus and thereby provide a “catalytic” surface that allows for the sequential processing of glycoproteins as they proceed through the ER and Golgi network.
  • the multiple compartments of the cis, medial, and trans Golgi and the trans-Golgi Network (TGN), provide the different localities in which the ordered sequence of glycosylation reactions can take place.
  • glycoprotein proceeds from synthesis in the ER to full maturation in the late Golgi or TGN, it is sequentially exposed to different glycosidases, mannosidases and glycosyltransferases such that a specific carbohydrate structure may be synthesized.
  • Much work has been dedicated to revealing the exact mechanism by which these enzymes are retained and anchored to their respective organelle.
  • the evolving picture is complex but evidence suggests that stem region, membrane spanning region and cytoplasmic tail, individually or in concert, direct enzymes to the membrane of individual organelles and thereby localize the associated catalytic domain to that locus (see, e.g., Gleeson (1998) Histochem. Cell Biol. 109:517-532).
  • GalT A full length GalT from humans was not even synthesized in yeast, despite demonstrably high transcription levels.
  • the transmembrane region of the same human GalT fused to an invertase reporter was able to direct localization to the yeast Golgi, albeit it at low production levels.
  • Schwientek and co-workers have shown that fusing 28 amino acids of a yeast mannosyltransferase (MNT1), a region containing a cytoplasmic tail, a transmembrane region and eight amino acids of the stem region, to the catalytic domain of human GalT are sufficient for Golgi localization of an active GalT.
  • MNT1 yeast mannosyltransferase
  • Other galactosyltransferases appear to rely on interactions with enzymes resident in particular organelles because, after removal of their transmembrane region, they are still able to localize properly.
  • Improper localization of a glycosylation enzyme may prevent proper functioning of the enzyme in the pathway.
  • Aspergillus nidulans which has numerous ⁇ -1,2-mannosidases (Eades and Hintz (2000) Gene 255(l):25-34), does not add GlcNAc to Man 5 GlcNAc 2 when transformed with the rabbit GnTI gene, despite a high overall level of GnTI activity (Kalsner et al. (1995) Glycoconj. J. 12(3):360-370).
  • GnTI although actively expressed, may be incorrectly localized such that the enzyme is not in contact with both of its substrates: UDP-GlcNAc and a productive Man 5 GlcNAc 2 substrate (not all Man 5 GlcNAc 2 structures are productive; see below).
  • the host organism may not provide an adequate level of UDP-GlcNAc in the Golgi or the enzyme may be properly localized but nevertheless inactive in its new environment.
  • Man 5 GlcNAc 2 structures present in the host cell may differ in structure from Man 5 GlcNAc 2 found in mammals. Maras and coworkers found that about one third of the N-glycans from cellobiohydrolase I (CBHI) obtained from T.
  • Man 5 GlcNAc 2 can be trimmed to Man 5 GlcNAc 2 by A. saitoi 1,2 mannosidase in vitro. Fewer than 1% of those N-glycans, however, could serve as a productive substrate for GnTI. Maras et al. (1997) Eur. J. Biochem. 249:701-707. The mere presence of Man 5 GlcNAc 2 , therefore, does not assure that further in vivo processing of Man 5 GlcNAc 2 can be achieved. It is formation of a productive, GnTI-reactive Man 5 GlcNAc 2 structure that is required. Although Man 5 GlcNAc 2 could be produced in the cell (about 27 mol %), only a small fraction could be converted to Man 5 GlcNAc 2 (less than about 5%, see Chiba et al. WO 01/14522).
  • glycosylation A significant number of proteins isolated from humans or animals are post-translationally modified, with glycosylation being one of the most significant modifications. An estimated 70% of all therapeutic proteins are glycosylated and thus currently rely on a production system (i.e., host cell) that is able to glycosylate in a manner similar to humans.
  • a production system i.e., host cell
  • glycosylation plays an important role in determining the (1) immunogenicity, (2) pharmacokinetic properties, (3) trafficking, and (4) efficacy of therapeutic proteins. It is thus not surprising that substantial efforts by the pharmaceutical industry have been directed at developing processes to obtain glycoproteins that are as “humanoid” or “human-like” as possible. To date, most glycoproteins are made in a mammalian host system.
  • This may involve the genetic engineering of such mammalian cells to enhance the degree of sialylation (i.e., terminal addition of sialic acid) of proteins expressed by the cells, which is known to improve pharmacokinetic properties of such proteins.
  • sialylation i.e., terminal addition of sialic acid
  • one may improve the degree of sialylation by in vitro addition of such sugars using known glycosyltransferases and their respective nucleotide sugars (e.g., 2,3-sialyltransferase and CMP-sialic acid).
  • glycoproteins produced in a microorganism host such as yeast utilizing the endogenous host glycosylation pathway differ structurally from those produced in mammalian cells and typically show greatly reduced therapeutic efficacy.
  • Such glycoproteins are typically immunogenic in humans and show a reduced half-life (and thus bioactivity) in vivo after administration (Takeuchi (1997) Trends in Glycoscience and Glycotechnology 9:S29-S35).
  • Specific receptors in humans and animals i.e., macrophage mannose receptors
  • Additional adverse effects may include changes in protein folding, solubility, susceptibility to proteases, trafficking, transport, compartmentalization, secretion, recognition by other proteins or factors, antigenicity, or allergenicity.
  • Yeast and filamentous fungi have both been successfully used for the production of recombinant proteins, both intracellular and secreted (Cereghino and Cregg (2000) FEMS Microbiology Reviews 24(1):45-66; Harkki et al. (1989) Bio - Technology 7(6):596; Berka et al. (1992) Abstr. Papers Amer. Chem. Soc. 203:121-BIOT; Svetina et al. (2000) J. Biotechnol. 76(2-3):245-251).
  • lactis, Pichia pastoris, Pichia methanolica, and Hansenula polymorpha have played particularly important roles as eukaryotic expression systems because they are able to grow to high cell densities and secrete large quantities of recombinant protein.
  • filamentous fungi such as Aspergillus niger, Fusarium sp, Neurospora crassa and others, have been used to efficiently produce glycoproteins at the industrial scale.
  • glycoproteins expressed in any of these eukaryotic microorganisms differ substantially in N-glycan structure from those in animals. This has prevented the use of yeast or filamentous fungi as hosts for the production of many therapeutic glycoproteins.
  • the first step the transfer of the core oligosaccharide structure to the nascent protein, is highly conserved in all eukaryotes including yeast, fungi, plants and humans (compare FIGS. 1A and 1B ).
  • Subsequent processing of the core oligosaccharide differs significantly in yeast and involves the addition of several mannose sugars.
  • This step is catalyzed by mannosyltransferases residing in the Golgi (e.g., OCH1, MNT1, MNN1, etc.), which sequentially add mannose sugars to the core oligosaccharide.
  • the resulting structure is undesirable for the production of humanoid proteins and it is thus desirable to reduce or eliminate mannosyltransferase activity.
  • Mutants of S. cerevisiae deficient in mannosyltransferase activity e.g., och1 or mnn9 mutants
  • Other oligosaccharide processing enzymes such as mannosylphosphate transferase, may also have to be eliminated depending on the host's particular endogenous glycosylation pattern.
  • the formation of complex N-glycans has to be engineered into the host system. This requires the stable expression of several enzymes and sugar-nucleotide transporters. Moreover, one has to localize these enzymes so that a sequential processing of the maturing glycosylation structure is ensured.
  • glycosyltransferases have been separately cloned and expressed in S. cerevisiae (GalT, GnTI), Aspergillus nidulans (GnTI) and other fungi (Yoshida et al. (1999) Glycobiology 9(1):53-8, Kalsner et al. (1995) Glycoconj. J 12(3):360-370).
  • GalT S. cerevisiae
  • GnTI Aspergillus nidulans
  • fungi Yoshida et al. (1999) Glycobiology 9(1):53-8, Kalsner et al. (1995) Glycoconj. J 12(3):360-370.
  • N-glycans resembling those made in human cells were not obtained.
  • Yeasts produce a variety of mannosyltransferases (e.g., 1,3-mannosyltransferases such as MNN1 in S. cerevisiae; Graham and Emr (1991) J. Cell. Biol. 114(2):207-218), 1,2-mannosyltransferases (e.g., KTR/KRE family from S. cerevisiae ), 1,6-mannosyltransferases (e.g., OCH1 from S. cerevisiae ), mannosylphosphate transferases and their regulators (e.g., MNN4 and MNN6 from S. cerevisiae ) and additional enzymes that are involved in endogenous glycosylation reactions.
  • 1,3-mannosyltransferases such as MNN1 in S. cerevisiae; Graham and Emr (1991) J. Cell. Biol. 114(2):207-218
  • 1,2-mannosyltransferases e.g., KTR/
  • Japanese Patent Application Publication No. 8-336387 discloses the deletion of an OCH1 homolog in Pichia pastoris.
  • OCH1 encodes a 1,6-mannosyltransferase, which adds a mannose to the glycan structure Man 8 GlcNAc 2 to yield Man 9 GlcNAc 2 .
  • the Man 9 GlcNAc 2 structure which contains three 1,6 mannose residues, is then a substrate for further 1,2-, 1,6-, and 1,3- mannosyltransferases in vivo, leading to the hypermannosylated glycoproteins that are characteristic for S. cerevisiae and which typically may have 30-40 mannose residues per N-glycan.
  • the Och1p initiates the transfer of 1,6 mannose to the Man 8 GlcNAc 2 core, it is often referred to as the “initiating 1,6 mannosyltransferase” to distinguish it from other 1,6 mannosyltransferases acting later in the Golgi.
  • the Och1p initiates the transfer of 1,6 mannose to the Man 8 GlcNAc 2 core, it is often referred to as the “initiating 1,6 mannosyltransferase” to distinguish it from other 1,6 mannosyltransferases acting later in the Golgi.
  • proteins glycosylated with Man 8 GlcNAc 2 accumulate and hypermannosylation does not occur.
  • Man 8 GlcNAc 2 is not a substrate for mammalian glycosyltransferases, such as human UDP-GlcNAc transferase I, and accordingly, the use of that mutant strain, in itself, is not useful for producing mammalian-like proteins, i.e., those with complex or hybrid glycosylation patterns.
  • Man 8 GlcNAc 2 structures to a Man 5 GlcNAc 2 isomer in S. cerevisiae (although high efficiency trimming greater than 50% in vivo has yet to be demonstrated) by engineering a fungal mannosidase from A. saitoi into the endoplasmic reticulum (ER).
  • ER endoplasmic reticulum
  • U.S. Pat. No. 5,834,251 discloses a method for producing a hybrid glycoprotein derived from Trichoderma reseei.
  • a hybrid N-glycan has only mannose residues on the Man ⁇ 1-6 arm of the core mannose structure and one or two complex antennae on the Man ⁇ 1-3 arm. While this structure has utility, the method has the disadvantage that numerous enzymatic steps must be performed in vitro, which is costly and time-consuming. Isolated enzymes are expensive to prepare and need costly substrates (e.g., UDP-GlcNAc). The method also does not allow for the production of complex glycans on a desired protein.
  • Alpha-1,2-mannosidase activity is required for the trimming of Man 8 GlcNAc 2 to form Man 5 GlcNAc 2 , which is a major intermediate for complex N-glycan formation in mammals.
  • Previous work has shown that truncated murine, fungal and human ⁇ -1,2-mannosidase can be expressed in the methylotropic yeast P. pastoris and display Man 8 GlcNAc 2 to Man 5 GlcNAc 2 trimming activity (Lal et al. (1998) Glycobiology 8(10):981-95; Tremblay et al. (1998) Glycobiology 8(6):585-95, Callewaert et al. (2001) FEBS Lett. 503(2-3):173-8).
  • Man 5 GlcNAc 2 While it is useful to engineer strains that are able to produce Man 5 GlcNAc 2 as the primary N-glycan structure, any attempt to further modify these high mannose precursor structures to more closely resemble human glycans requires additional in vivo or in vitro steps. Methods to further humanize glycans from fungal and yeast sources in vitro are described in U.S. Pat. No. 5,834,251 (supra). If Man 5 GlcNAc 2 is to be further humanized in vivo, one has to ensure that the generated Man 5 GlcNAc 2 structures are, in fact, generated intracellularly and not the product of mannosidase activity in the medium.
  • glycoproteins characterized by a high intracellular Man 5 GlcNAc 2 content which can be further processed into human-like glycoprotein structures in non-human eukaryotic host cells, and particularly in yeast and filamentous fungi.
  • GnTs N-Acetylglucosaminyltransferases
  • GnTs N-Acetylglucosaminyltransferases
  • Such glycosyltransferases catalyze the transfer of a monosaccharide from specific sugar nucleotide donors onto particular hydroxyl position of a monosaccharide in a growing glycan chain in one of two possible anomeric linkages (either ⁇ or ⁇ ).
  • GnTs add N-acetylglucosamine (“GlcNAc”) onto the Man ⁇ 1,6 arm or the Man ⁇ 1,3 arm of an N-glycan substrate (e.g., Man 5 GlcNAc 2 (“mannose-5 core”) and Man 3 GlcNAc 2 (an “inner core structure”)).
  • the reaction product e.g., GlcNAcMan 5 GlcNAc 2 or GlcNAc 2 Man 3 GlcNAc 2
  • GnTIII N-Acetylglucosaminyltransferase III
  • a GlcNAc is an enzyme that catalyzes the addition of a GlcNAc, on the middle mannose of the trimannose core (Man ⁇ 1,6 (Man ⁇ 1,3) Man ⁇ 1,4-GlcNAc ⁇ 1,4-GlcNAc ⁇ 1,4-Asn) of an N-linked oligosaccharide.
  • the addition by GnTIII of a bisecting GlcNAc to an acceptor substrate e.g. trimannose core
  • the addition by GnTIII of a bisecting GlcNAc to the GlcNAcMan 3 GlcNAc 2 structure may yield a bisected N-glycan, GlcNAc 2 Man 3 GlcNAc 2 .
  • the addition by GnTIII of a bisecting GlcNAc to a GlcNAc 2 Man 3 GlcNAc 2 structure yields another bisected N-glycan, GlcNAc 3 Man 3 GlcNAc 2 .
  • This latter structure has been implicated in greater antibody-dependent cellular cytotoxicity (ADCC). Umana et al. (1999) Nat. Biotechnol. 17(2): 176-80.
  • GlcNAcMan 4 GlcNAc 2 can be converted to bisected GlcNAc 2 Man 4 GlcNAC 2
  • Man 5 GlcNAc 2 can be converted to bisected GlcNAcMan 5 GlcNAc 2
  • GlcNAcMan 5 GlcNAc 2 can be converted to bisected GlcNAc 2 Man 5 GlcNAc 2 .
  • Narasimhan (1982) J. Biol. Chem. 257:10235-42 See, e.g., Narasimhan (1982) J. Biol. Chem. 257:10235-42.
  • GnTIII activity has only been shown in mammalian cells.
  • Host cells of the invention e.g., lower eukaryotic microorganisms and other non-human, eukaryotic host cells grown in culture, are modified to produce N-glycans, such as bisected N-glycans, or other structures produced along human glycosylation pathways.
  • the present invention thus provides a glycoprotein production method using (1) a lower eukaryotic host such as a unicellular or filamentous fungus, or (2) any non-human eukaryotic organism that has a different glycosylation pattern from humans, to modify the glycosylation composition and structures of the proteins made in a host organism (“host cell”) so that they resemble more closely carbohydrate structures found in mammalian, e.g., human proteins.
  • host cell a host organism
  • the process allows one to obtain an engineered host cell which can be used to express and target any desirable gene(s), e.g., one involved in glycosylation, by methods that are well-established in the scientific literature and generally known to the artisan in the field of protein expression.
  • Host cells with modified oligosaccharides are created or selected. For the production of therapeutic proteins, this method may be adapted to engineer cell lines in which any desired glycosylation structure may be obtained.
  • the invention provides methods for making a human-like glycoprotein in a lower eukaryotic host cell by introduction into the cell of an N-acetylglucosaminyltransferase III activity.
  • the N-acetylglucosaminyltransferase III activity is expressed in the cell, and in an even more preferred embodiment, this expression results in the production of N-glycans comprising GlcNAc 3 Man 3 GlcNAc 2 , GlcNAc 2 Man 3 GlcNAc 2 , or GlcNAc 2 Man 5 GlcNAc 2 bisected structures.
  • the N-acetylglucosaminyltransferase III activity is substantially intracellular.
  • the glycoprotein including the N-glycans with bisected structures is isolated from the lower eukaryotic host cell.
  • the glycoprotein produced in the host cell is a therapeutic protein.
  • the invention provides a lower eukaryotic host cell that includes both an N-acetylglucosaminyltransferase III activity and an N-acetylglucosaminyltransferase II activity.
  • the host cell including the N-acetylglucosaminyltransferase III activity produces N-glycans comprising GlcNAcMan 3 GlcNAc 2 structures that are capable of reacting with this activity.
  • the activity produces a bisected glycan.
  • the lower eukaryotic host cell of some embodiments of the invention may thus include an N-glycan with a bisected glycan.
  • the N-glycan includes greater than 10 mole % of the bisected glycan.
  • the host cell includes an N-glycan that comprises GlcNAc 3 Man 3 GlcNAc 2 , GlcNAc 2 Man 3 GlcNAc 2 , or GlcNAc 2 Man 5 GlcNAc 2 bisected structures.
  • the host cell includes a Man 5 GlcNAc 2 core structure or a Man 3 GlcNAc 2 core structure that is modified by a bisecting GlcNAc.
  • the cell produces greater than 10 mole % of the modified structure.
  • the lower eukaryotic host cell contains an N-acetylglucosaminyltransferase I activity in addition to the N-acetylglucosaminyltransferase III activity.
  • the activities are substantially intracellular.
  • the cell produces N-glycans comprising GlcNAcMan 3 GlcNAc 2 that are capable of reacting with the GnTIII activity.
  • the GnTIII activity of the cell produces a bisected glycan.
  • the lower eukaryotic host cell of the invention contains both an N-acetylglucosaminyltransferase III activity and a mannosidase II activity.
  • the host cell further contains an N-acetylglucosaminyltransferase I activity.
  • the host cell further contains an N-acetylglucosaminyltransferase II activity.
  • the host cell further contains both an N-acetylglucosaminyltransferase I activity and an N-acetylglucosaminyltransferase II activity.
  • the host cell of the invention is deficient in an OCH1 mannosyltransferase activity.
  • Such a cell may, for example, be deficient in a Do1-P-Man:Man5GlcNAc2-PP-Do1 mannosyltransferase activity.
  • the host cell of the invention may further comprise an ⁇ -1,2-mannosidase I activity.
  • the host cell may further comprise a UDP-GlcNAc transporter.
  • the present invention also provides glycoproteins that are made by the processes of the invention.
  • the glycoprotein includes a bisecting GlcNAc on a Man 5 GlcNAc 2 or a Man 3 GlcNAc 2 core structure and is produced in a lower eukaryotic host cell.
  • the glycoprotein includes a bisecting GlcNAc attached to a Man 5 GlcNAc 2 , Man 4 GlcNAc 2 .
  • Man 3 GlcNAc 2 Man 3 GlcNAc 2 , GlcNAcMan 3 GlcNAc 2 , GlcNAcMan 5 GlcNAc 2 , or a GlcNAc 2 Man 3 GlcNAc 2 core structure and is produced in a lower eukaryotic host cell.
  • greater than 10 mole % of the core structures of the glycoprotein of the invention are modified by the bisecting GlcNAc.
  • the invention provides pharmaceutical compositions that contain the human-like glycoproteins produced in a lower eukaryotic host cell.
  • vectors encoding proteins having N-acetylglucosaminyltransferase III activity and containing attached targeting peptide sequences.
  • the proteins encoded by the vectors are localized in a lower eukaryotic host cell such that they produce N-glycans having bisected structures.
  • FIG. 1A is a schematic diagram of a typical fungal N-glycosylation pathway.
  • FIG. 1B is a schematic diagram of a typical human N-glycosylation pathway.
  • FIG. 2 depicts construction of a combinatorial DNA library of fusion constructs.
  • FIG. 2A diagrams the insertion of a targeting peptide fragment into pCR2.1-TOPO (Invitrogen, Carlsbad, Calif.).
  • FIG. 2B shows the generated targeting peptide sub-library having restriction sites NotI-AscI.
  • FIG. 2C diagrams the insertion of a catalytic domain region into pJN347, a modified pUC19 vector.
  • FIG. 2D shows the generated catalytic domain sub-library having restriction sites NotI, AscI and PacI.
  • FIG. 2E depicts one particular fusion construct generated from the targeting peptide sub-library and the catalytic domain sub-library.
  • FIG. 3 illustrates the M. musculus ⁇ -1,2-mannosidase IA open reading frame nucleic acid sequence (SEQ ID NO:48) and encoded polypeptide sequence (SEQ ID NO:49). The sequences of the PCR primers used to generate N-terminal truncations are underlined.
  • FIG. 4 illustrates engineering of vectors with multiple auxotrophic markers and genetic integration of target proteins in the P. pastoris OCH1 locus.
  • FIGS. 5A-5E show MALDI-TOF analysis demonstrating production of kringle 3 domain of human plasminogen (K3) glycoproteins having Man 5 GlcNAc 2 as the predominant N-glycan structure in P. pastoris.
  • FIG. 5A depicts the standard Man 5 GlcNAc 2 [a] glycan (Glyko, Novato, Calif.) and Man 5 GlcNAc 2 +Na + [b].
  • FIG. 5B shows PNGase—released glycans from K3 wild type.
  • FIG. 5C depicts the och1 deletion resulting in the production of Man 8 GlcNAc 2 [c] as the predominant N-glycan.
  • FIGS. 5D and 5E show the production of Man 5 GlcNAc 2 [b] after in vivo trimming of Man 8 GlcNAc 2 with a chimeric ⁇ -1,2-mannosidase.
  • the predominant N-glycan is indicated by a peak with a mass (m/z) of 1253 consistent with its identification as Man 5 GlcNAc 2 [b].
  • FIGS. 6A-6F show MALDI-TOF analysis demonstrating production of IFN- ⁇ glycoproteins having Man 5 GlcNAc 2 as the predominant N-glycan structure in P. pastoris.
  • FIG. 6A shows the standard Man 5 GlcNAc 2 [a] and Man 5 GlcNAc 2 +Na + [b] as the standard (Glyko, Novato, Calif.).
  • FIG. 6B shows PNGase—released glycans from IFN- ⁇ wildtype.
  • FIG. 6C depicts the och1 knock-out producing Man 8 GlcNAc 2 [c]; Man 9 GlcNAc 2 [d]; Man 10 GlcNAc 2 [e]; Man 11 GlcNAc 2 [f]; Man 12 GlcNAc 2 [g]; and no production of Man 5 GlcNAc 2 [b].
  • FIG. 6D shows relatively small amount of Man 5 GlcNAc 2 [b] among other intermediate N-glycans Man 8 GlcNAc 2 [c] to Man 12 GlcNAc 2 [g].
  • FIG. 6E shows a significant amount of Man 5 GlcNAc 2 [b] relative to the other glycans Man 8 GlcNAc 2 [c] and Man 9 GlcNAc 2 [d] produced by pGC5 ( Saccharomyces MNS1(m)/mouse mannosidase IB ⁇ 99).
  • FIG. 6F shows predominant production of Man 5 GlcNAc 2 [b] on the secreted glycoprotein IFN- ⁇ by pFB8 ( Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187).
  • the N-glycan is indicated by a peak with a mass (m/z) of 1254 consistent with its identification as Man 5 GlcNAc 2 [b].
  • FIG. 7 shows a high performance liquid chromatogram for: (A) Man 9 GlcNAc 2 standard labeled with 2-AB (negative control); (B) supernatant of medium P. pastoris, ⁇ och1 transformed with pFB8 mannosidase, which demonstrates a lack of extracellular mannosidase activity in the supernatant; and (C) Man 9 GlcNAc 2 standard labeled with 2-AB after exposure to T. reesei mannosidase (positive control).
  • FIG. 8 shows a high performance liquid chromatogram for: (A) Man 9 GlcNAc 2 standard labeled with 2-AB (negative control); (B) supernatant of medium P. pastoris, ⁇ och1 transformed with pGC5 mannosidase, which demonstrates a lack of extracellular mannosidase activity in the supernatant; and (C) Man 9 GlcNAc 2 standard labeled with 2-AB after exposure to T. reesei mannosidase (positive control).
  • FIG. 9 shows a high performance liquid chromatogram for: (A) Man 9 GlcNAc 2 standard labeled with 2-AB (negative control); (B) supernatant of medium P. pastoris, ⁇ och1 transformed with pBC18-5 mannosidase, which demonstrates lack of extracellular mannosidase activity in the supernatant; and (C) supernatant of medium P. pastoris, ⁇ och1 transformed with pDD28-3, which demonstrates activity in the supernatant (positive control).
  • FIGS. 10A-10B demonstrate the activity of an UDP-GlcNAc transporter in the production of GlcNAcMan 5 GlcNAc 2 in P. pastoris.
  • FIG. 10A depicts a P. pastoris strain (YSH-3) with a human GnTI but without the UDP-GlcNAc transporter resulting in some production of GlcNAcMan 5 GlcNAc 2 [b] but a predominant production of Man 5 GlcNAc 2 [a].
  • FIG. 10B depicts the addition of UDP-GlcNAc transporter from K. lactis in a strain (PBP-3) with the human GnTI, which resulted in the predominant production of GlcNAcMan 5 GlcNAc 2 [b].
  • the single prominent peak of mass (m/z) at 1457 is consistent with its identification as GlcNAcMan 5 GlcNAc 2 [b] as shown in FIG. 10B .
  • FIG. 11 shows a pH optimum of a heterologous mannosidase enzyme encoded by pBB27-2 ( Saccharomyces MNN10 (s)/ C. elegans mannosidase IB ⁇ 31) expressed in P. pastoris.
  • pBB27-2 Saccharomyces MNN10 (s)/ C. elegans mannosidase IB ⁇ 31
  • FIGS. 12A-12C show MALDI-TOF analysis of N-glycans released from a cell free extract of K. lactis.
  • FIG. 12A shows the N-glycans released from wild-type cells, which includes high-mannose type N-glycans.
  • FIG. 12B shows the N-glycans released from och1 mnn1 deleted cells, revealing a distinct peak of mass (m/z) at 1908 consistent with its identification as Man 9 GlcNAc 2 [d].
  • FIG. 12C shows the N-glycans released from ochl mnnl deleted cells after in vitro ⁇ -1,2-mannosidase digest corresponding to a peak consistent with Man 5 GlcNAc 2 .
  • FIG. 13 is a schematic of the structure of the dolichyl pyrophosphate-linked oligosaccharide.
  • FIG. 14 is a schematic of the generation of GlcNAc 2 Man 3 GlcNAc 2 N-glycans from fungal host cells which are deficient in alg3, alg9, or alg12 activities.
  • FIG. 15 is a schematic of processing reactions required to produce mammalian-type oligosaccharide structures in a fungal host cell with an alg3, och1 genotype.
  • FIG. 16 shows S. cerevisiae Alg3 Sequence Comparisons (Blast) (SEQ ID NOs:9-20, respectively, in order of appearance)
  • FIG. 17 shows S. cerevisiae ALG3 (SEQ ID NO:21) and Alg3p (SEQ ID NO:22) Sequences
  • FIG. 18 shows P. pastoris ALG3 (SEQ ID NO:23) and Alg3p (SEQ ID NO:24) Sequences
  • FIG. 19 shows P. pastoris ALG3 Sequence Comparisons (Blast) (SEQ ID NOs:23-31, respectively, in order of appearance)
  • FIG. 20 shows K. lactis ALG3 (SEQ ID NO:33) and Alg3p (SEQ ID NO:34) Sequences
  • FIG. 21 shows K. lactis ALG3 Sequence Comparisons (Blast) (SEQ ID NOs:35-40, respectively, in order of appearance)
  • FIG. 22 shows a model of an IgG immunoglobulin.
  • Heavy chain and light chain can be, based on similar secondary and tertiary structure, subdivided into domains.
  • the two heavy chains (domains V H , C H 1, C H 2 and C H 3) are linked through three disulfide bridges.
  • the light chains (domains V L and C L ) are linked by another disulfide bridge to the C H 1 portion of the heavy chain and, together with the C H 1 and V H fragments, make up the Fab region.
  • Antigens bind to the terminal portion of the Fab region. Effector-functions, such as Fc-gamma-Receptor binding have been localized to the C H 2 domain, just downstream of the hinge region and are influenced by N-glycosylation of asparagine 297 in the heavy chain.
  • FIG. 23 is a schematic overview of a modular IgG1 expression vector.
  • FIG. 24 shows M. musculis GnTIII Nucleic Acid (SEQ ID NO:45) And Amino Acid (SEQ ID NO:46) Sequences
  • FIG. 25 (top) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in a P. pastoris YSH-1 displaying a predominant peak at 1461 m/z corresponding to the the mass of GlcNAcMan 5 GlcNAc 2 [d];
  • FIG. 25 (bottom) shows a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in a P. pastoris YSH-1 transformed with D. melanogaster mannosidase II ⁇ 74/ S.
  • FIG. 26 (top) is the MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in P. pastoris YSH-1 as shown in FIG. 25 (top);
  • FIG. 26 (bottom) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-1 cells transformed with a pVA53 construct ( S. cerevisiae MNN2(s)/mGnTIII).
  • the peak at 1463 m/z corresponds the mass of GlcNAcMan 5 GlcNAc 2 [d] and the peak at 1666 m/z corresponds to the mass of GlcNAc 2 Man 5 GlcNAc 2 [a].
  • FIG. 27 (top) is the MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in P. pastoris YSH-1 as shown in FIG. 25 (top);
  • FIG. 27 (bottom) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-1 cells transformed with a pVA55 construct ( S. cerevisiae MNN2(s)/mGnTIII).
  • the peak at 1463 m/z corresponds to the mass of GlcNAcMan 5 GlcNAc 2 [d] and the peak at 1667 m/z corresponds to the mass of GlcNAc 2 Man 5 GlcNAc 2 [a].
  • FIG. 28 (top) is the MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in P. pastoris YSH-1 as shown in FIG. 25 (top);
  • FIG. 28 (bottom) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-1 cells transformed with a pVB51 construct ( K. lactis GNT1(s)/mGnTIII).
  • the predominant peak at 1463 m/z corresponds to the mass of GlcNAcMan 5 GlcNAc 2 [d] and a second peak at 1726 m/z [e], which does not correpond to the mass of GlcNAc 2 Man 5 GlcNAc 2 is observed.
  • FIG. 29 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-44 cells.
  • the predominant peak at 1356 m/z corresponds to the mass of GlcNAc 2 Man 3 GlcNAc 2 [x].
  • FIG. 30 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-44 cells transformed with a pVA53 construct ( S. cerevisiae MAN2(s)/mGnTIII).
  • the peak at 1340 m/z corresponds to the mass of GlcNAc 2 Man 3 GlcNAc 2 [x] and the peak at 1542 m/z corresponds to the mass of GlcNAc 3 Man 3 GlcNAc 2 [y].
  • FIG. 31 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris PBP6-5 cells.
  • the predominant peak at 1340 m/z corresponds to the mass of GlcNAc 2 Man 3 GlcNAc 2 [X].
  • FIG. 32 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris PBP6-5 cells transformed with a pVA53 construct ( S. cerevisiae MNN2(s)/mGnTIII).
  • the peak at 1340 m/z corresponds to the mass of GlcNAc 2 Man 3 GlcNAc 2 [x] and the peak at 1543 m/z correponds to the mass of GlcNAc 3 Man 3 GlcNAc 2 [y].
  • FIG. 33 shows a high performance liquid chromatogram, which demonstrates a lack of extracellular GnTIII activity (pVA53) in the supernatant.
  • the N-glycan GlcNAcMan 5 GlcNAc 2 purified from K3 expressed in PBP-3 strain was added to: BMMY (A); 1 mM UDP-GlcNAc (Sigma Chemical Co., St. Louis, Mo.)) in BMMY (B); the supernatant of YSH-44 transformed with pVA53 [YSH-57] (C); and the supernatant of YSH-57+1 mM UDP-GlcNAc (D).
  • FIG. 34 shows a high performance liquid chromatogram, which demonstrates a lack of extracellular GnTIII activity (pVA53) in the supernatant.
  • the N-glycan GlcNAc 2 Man 3 GlcNAc 2 purified from K3 expressed in YSH-44 strain was added to: BMMY (A); 1 mM UDP-GlcNAc (Sigma Chemical Co., St. Louis, Mo.)) in BMMY (B); and the supernatant of YSH-44 transformed with pVA53 [YSH-57] (C).
  • FIG. 35 is a schematic diagram comparing the normal glycosylation pathways in humans and P. pastoris (Panel A) with an engineered humanized N-glycosylation pathway in lower eukaryotes (Panel B).
  • the engineered pathway represents the construction of P. pastoris strain PBP6-5, which after modification with GnTIII becomes P. pastoris strain PBP38.
  • FIG. 36 is a schematic diagram showing the predominant secreted glycoform produced by each of the designated P. pastoris strains and the gene modification used to engineer each of the strains.
  • FIG. 37 is a structural representation of the transfer of a GlcNAc to the oligosaccharide intermediate, GlcNAcMan 5 GlcNAc 2 , produced on glycoproteins in a lower eukaryotic host cell, as catalyzed by GnTIII.
  • FIG. 38 is a structural representation of the transfer of a GlcNAc to the oligosaccharide intermediate, GlcNAcMan 3 GlcNAc 2 , produced on glycoproteins in a lower eukaryotic host cell, as catalyzed by GnTII, and the subsequent transfer of a GlcNAc to the product of that reaction, GlcNAc 2 Man 3 GlcNAc 2 , as catalyzed by GnTIII.
  • N-glycan refers to an N-linked oligosaccharide, e.g., one that is attached by an asparagine-N-acetylglucosamine linkage to an asparagine residue of a polypeptide.
  • N-glycans have a common pentasaccharide core of Man 3 GlcNAc 2 (“Man” refers to mannose; “Glc” refers to glucose; and “NAc” refers to N-acetyl; GlcNAc refers to N-acetylglucosamine).
  • Mannose core used with respect to the N-glycan also refers to the structure Man 3 GlcNAc 2 (“Man 3 ”).
  • N-glycan refers to the structure Man 5 GlcNAc 2 .
  • N-glycans differ with respect to the number of branches (antennae) comprising peripheral sugars (e.g., GlcNAc, fucose, and sialic acid) that are attached to the Man 3 core structure.
  • branches e.g., g., GlcNAc, fucose, and sialic acid
  • N-glycans are classified according to their branched constituents (e.g., high mannose, complex or hybrid).
  • a “high mannose” type N-glycan has five or more mannose residues.
  • a “complex” type N-glycan typically has at least one GlcNAc attached to the 1,3 mannose arm and at least one GlcNAc attached to the 1,6 mannose arm of the trimannose core.
  • Complex N-glycans may also have galactose (“Gal”) residues that are optionally modified with sialic acid or derivatives (“NeuAc”, where “Neu” refers to neuraminic acid and “Ac” refers to acetyl).
  • a complex N-glycan typically has at least one branch that terminates in an oligosaccharide such as, for example: NeuNAc-; NeuAc ⁇ 2-6GalNAc ⁇ 1-; NeuAc ⁇ 2-3Gal ⁇ 1-3GalNAc ⁇ 1-; NeuAc ⁇ 2-3/6Gal ⁇ 1-4GlcNAc ⁇ 1-; GlcNAc ⁇ 1-4Gal ⁇ 1-(mucins only); Fuc ⁇ 1-2Gal ⁇ 1-(blood group H).
  • Sulfate esters can occur on galactose, GalNAc, and GlcNAc residues, and phosphate esters can occur on mannose residues.
  • NeuAc Neuro: neuraminic acid; Ac:acetyl
  • NeuG1 N-glycolylneuraminic acid
  • Complex N-glycans may also have intrachain substitutions comprising “bisecting” GlcNAc and core fucose (“Fuc”).
  • a “hybrid” N-glycan has at least one GlcNAc on the terminal of the 1,3 mannose arm of the trimannose core and zero or more mannoses on the 1,6 mannose arm of the trimannose core.
  • N-glycans refers to a structure which represents the major peak detected by matrix assisted laser desorption ionization time of flight mass spectrometry (MALDI-TOF) analysis.
  • MALDI-TOF matrix assisted laser desorption ionization time of flight mass spectrometry
  • Abbreviations used herein are of common usage in the art, see, e.g., abbreviations of sugars, above.
  • Other common abbreviations include “PNGase”, which refers to peptide N-glycosidase F (EC 3.2.2.18); “GlcNAc Tr” or “GnT,” which refers to N-acetylglucosaminyl Transferase enzymes; “NANA” refers to N-acetylneuraminic acid.
  • a “humanized glycoprotein” or a “human-like glycoprotein” refers alternatively to a protein having attached thereto N-glycans having fewer than four mannose residues, and synthetic glycoprotein intermediates (which are also useful and can be manipulated further in vitro or in vivo) having at least five mannose residues.
  • glycoproteins produced according to the invention contain at least 30 mole %, preferably at least 40 mole % and more preferably 50, 60, 70, 80, 90, or even 100 mole % of the Man 5 GlcNAc 2 intermediate, at least transiently.
  • a host cell of the invention may express a “better”, i.e., a more efficient glycosylation enzyme.
  • a mannosidase is selected such that it will have optimal activity under the conditions present at the site in the host cell where proteins are glycosylated and is introduced into the host cell preferably by targeting the enzyme to a host cell organelle where activity is desired.
  • enzyme when used herein in connection with altering host cell glycosylation, refers to a molecule having at least one enzymatic activity, and includes full-length enzymes, catalytically active fragments, chimerics, complexes, and the like.
  • a “catalytically active fragment” of an enzyme refers to a polypeptide having a detectable level of functional (enzymatic) activity.
  • Enzyme activity is “substantially intracellular” when less than 10% of the enzyme activity is measurable outside the cell compared to that measurable from lysed cells.
  • a lower eukaryotic host cell when used herein in connection with glycosylation profiles, refers to any eukaryotic cell which ordinarily produces high mannose containing N-glycans, and thus is meant to include some animal or plant cells and most typical lower eukaryotic cells, including uni- and multicellular fungal and algal cells.
  • secretion pathway refers to the assembly line of various glycosylation enzymes to which a lipid-linked oligosaccharide precursor and an N-glycan substrate are sequentially exposed, following the molecular flow of a nascent polypeptide chain from the cytoplasm to the endoplasmic reticulum (ER) and the compartments of the Golgi apparatus. Enzymes are said to be localized along this pathway.
  • An enzyme X that acts on a lipid-linked glycan or an N-glycan before enzyme Y is said to be or to act “upstream” to enzyme Y; similarly, enzyme Y is or acts “downstream” from enzyme X.
  • targeting peptide refers to nucleotide or amino acid sequences encoding a cellular targeting signal peptide which mediates the localization. (or retention) of an associated sequence to sub-cellular locations, e.g., organelles.
  • nucleic acid molecule refers to a polymeric form of nucleotides of at least 10 bases in length.
  • the term includes DNA molecules (e.g., cDNA or genomic or synthetic DNA) and RNA molecules (e.g., mRNA or synthetic RNA), as well as analogs of DNA or RNA containing non-natural nucleotide analogs, non-native internucleoside bonds, or both.
  • the nucleic acid can be in any topological conformation.
  • the nucleic acid can be single-stranded, double-stranded, triple-stranded, quadruplexed, partially double-stranded, branched, hairpinned, circular, or in a padlocked conformation.
  • the term includes single and double stranded forms of DNA.
  • a nucleic acid molecule of this invention may include both sense and antisense strands of RNA, cDNA, genomic DNA, and synthetic forms and mixed polymers of the above. They may be modified chemically or biochemically or may contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those of skill in the art.
  • Such modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen, etc.), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids, etc.)
  • synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen bonding and other chemical interactions. Such molecules are known in the art and include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
  • nucleic acid comprising SEQ ID NO:X refers to a nucleic acid, at least a portion of which has either (i) the sequence of SEQ ID NO:X, or (ii) a sequence complementary to SEQ ID NO:X.
  • the choice between the two is dictated by the context. For instance, if the nucleic acid is used as a probe, the choice between the two is dictated by the requirement that the probe be complementary to the desired target.
  • nucleic acid or polynucleotide e.g., an RNA, DNA or a mixed polymer
  • an “isolated” or “substantially pure” nucleic acid or polynucleotide is one which is substantially separated from other cellular components that naturally accompany the native polynucleotide in its natural host cell, e.g., ribosomes, polymerases, and genomic sequences with which it is naturally associated.
  • the term embraces a nucleic acid or polynucleotide that (1) has been removed from its naturally occurring environment, (2) is not associated with all or a portion of a polynucleotide in which the “isolated polynucleotide” is found in nature, (3) is operatively linked to a polynucleotide which it is not linked to in nature, or (4) does not occur in nature.
  • isolated or substantially pure also can be used in reference to recombinant or cloned DNA isolates, chemically synthesized polynucleotide analogs, or polynucleotide analogs that are biologically synthesized by heterologous systems.
  • isolated does not necessarily require that the nucleic acid or polynucleotide so described has itself been physically removed from its native environment.
  • an endogenous nucleic acid sequence in the genome of an organism is deemed “isolated” herein if a heterologous sequence (i.e., a sequence that is not naturally adjacent to this endogenous nucleic acid sequence) is placed adjacent to the endogenous nucleic acid sequence, such that the expression of this endogenous nucleic acid sequence is altered.
  • a non-native promoter sequence can be substituted (e.g., by homologous recombination) for the native promoter of a gene in the genome of a human cell, such that this gene has an altered expression pattern. This gene would now become “isolated” because it is separated from at least some of the sequences that naturally flank it.
  • a nucleic acid is also considered “isolated” if it contains any modifications that do not naturally occur to the corresponding nucleic acid in a genome.
  • an endogenous coding sequence is considered “isolated” if it contains an insertion, deletion or a point mutation introduced artificially, e.g., by human intervention.
  • An “isolated nucleic acid” also includes a nucleic acid integrated into a host cell chromosome at a heterologous site, a nucleic acid construct present as an episome.
  • an “isolated nucleic acid” can be substantially free of other cellular material, or substantially free of culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • the phrase “degenerate variant” of a reference nucleic acid sequence encompasses nucleic acid sequences that can be translated, according to the standard genetic code, to provide an amino acid sequence identical to that translated from the reference nucleic acid sequence.
  • sequence identity refers to the residues in the two sequences which are the same when aligned for maximum correspondence.
  • the length of sequence identity comparison may be over a stretch of at least about nine nucleotides, usually at least about 20 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36 or more nucleotides.
  • polynucleotide sequences can be compared using FASTA, Gap or Bestfit, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wis.
  • FASTA provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson (1990) Methods Enzymol. 183:63-98, incorporated herein by reference in its entirety).
  • percent sequence identity between nucleic acid sequences can be determined using FASTA with its default parameters (a word size of 6 and the NOPAM factor for the scoring matrix) or using Gap with its default parameters as provided in GCG Version 6.1, herein incorporated by reference.
  • nucleic acid or fragment thereof indicates that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 50%, more preferably 60% of the nucleotide bases, usually at least about 70%, more usually at least about 80%, preferably at least about 90%, and more preferably at least about 95%, 96%, 97%, 98% or 99% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or Gap, as discussed above.
  • nucleic acid or fragment thereof hybridizes to another nucleic acid, to a strand of another nucleic acid, or to the complementary strand thereof, under stringent hybridization conditions.
  • Stringent hybridization conditions and “stringent wash conditions” in the context of nucleic acid hybridization experiments depend upon a number of different physical parameters. Nucleic acid hybridization will be affected by such conditions as salt concentration, temperature, solvents, the base composition of the hybridizing species, length of the complementary regions, and the number of nucleotide base mismatches between the hybridizing nucleic acids, as will be readily appreciated by those skilled in the art. One having ordinary skill in the art knows how to vary these parameters to achieve a particular stringency of hybridization.
  • “stringent hybridization” is performed at about 25° C. below the thermal melting point (T m ) for the specific DNA hybrid under a particular set of conditions.
  • “Stringent washing” is performed at temperatures about 5° C. lower than the T m for the specific DNA hybrid under a particular set of conditions.
  • the T m is the temperature at which 50% of the target sequence hybridizes to a perfectly matched probe. See Sambrook et al., supra, page 9.51, hereby incorporated by reference.
  • high stringency conditions are defined for solution phase hybridization as aqueous hybridization (i.e., free of formamide) in 6 ⁇ SSC (where 20 ⁇ SSC contains 3.0 M NaCl and 0.3 M sodium citrate), 1% SDS at 65° C. for 8-12 hours, followed by two washes in 0.2 ⁇ SSC, 0.1% SDS at 65° C. for 20 minutes. It will be appreciated by the skilled artisan that hybridization at 65° C. will occur at different rates depending on a number of factors including the length and percent identity of the sequences which are hybridizing.
  • mutated when applied to nucleic acid sequences means that nucleotides in a nucleic acid sequence may be inserted, deleted or changed compared to a reference nucleic acid sequence. A single alteration may be made at a locus (a point mutation) or multiple nucleotides may be inserted, deleted or changed at a single locus. In addition, one or more alterations may be made at any number of loci within a nucleic acid sequence.
  • a nucleic acid sequence may be mutated by any method known in the art including but not limited to mutagenesis techniques such as “error-prone PCR” (a process for performing PCR under conditions where the copying fidelity of the DNA polymerase is low, such that a high rate of point mutations is obtained along the entire length of the PCR product. See, e.g., Leung et al. (1989) Technique 1:11-15 and Caldwell and Joyce (1992) PCR Methods Applic. 2:28-33); and “oligonucleotide-directed mutagenesis” (a process which enables the generation of site-specific mutations in any cloned DNA segment of interest. See, e.g., Reidhaar-Olson et al. (1988) Science 241:53-57).
  • mutagenesis techniques such as “error-prone PCR” (a process for performing PCR under conditions where the copying fidelity of the DNA polymerase is low, such that a high rate of point mutations is obtained along the
  • vector as used herein is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • Other vectors include cosmids, bacterial artificial chromosomes (BAC) and yeast artificial chromosomes (YAC).
  • BAC bacterial artificial chromosome
  • YAC yeast artificial chromosome
  • viral vector Another type of vector, wherein additional DNA segments may be ligated into the viral genome (discussed in more detail below).
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., vectors having an origin of replication which functions in the host cell).
  • vectors can be integrated into the genome of a host cell upon introduction into the host cell, and are thereby replicated along with the host genome. Moreover, certain preferred vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
  • “Operatively linked” expression control sequences refers to a linkage in which the expression control sequence is contiguous with the gene of interest to control the gene of interest, as well as expression control sequences that act in trans or at a distance to control the gene of interest.
  • expression control sequence refers to polynucleotide sequences which are necessary to affect the expression of coding sequences to which they are operatively linked. Expression control sequences are sequences which control the transcription, post-transcriptional events and translation of nucleic acid sequences. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (e.g., ribosome binding sites); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion.
  • control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence.
  • control sequences is intended to include, at a minimum, all components whose presence is essential for expression, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • recombinant host cell (or simply “host cell”), as used herein, is intended to refer to a cell into which a nucleic acid such as a recombinant vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • a recombinant host cell may be an isolated cell or cell line grown in culture or may be a cell which resides in a living tissue or organism.
  • peptide refers to a short polypeptide, e.g., one that is typically less than about 50 amino acids long and more typically less than about 30 amino acids long.
  • the term as used herein encompasses analogs and mimetics that mimic structural and thus biological function.
  • polypeptide as used herein encompasses both naturally-occurring and non-naturally-occurring proteins, and fragments, mutants, derivatives and analogs thereof.
  • a polypeptide may be monomeric or polymeric. Further, a polypeptide may comprise a number of different domains each of which has one or more distinct activities.
  • isolated protein or “isolated polypeptide” is a protein or polypeptide that by virtue of its origin or source of derivation (1) is not associated with naturally associated components that accompany it in its native state, (2) when it exists in a purity not found in nature, where purity can be adjudged with respect to the presence of other cellular material (e.g., is free of other proteins from the same species) (3) is expressed by a cell from a different species, or (4) does not occur in nature (e.g., it is a fragment of a polypeptide found in nature or it includes amino acid analogs or derivatives not found in nature or linkages other than standard peptide bonds).
  • polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components.
  • a polypeptide or protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well-known in the art.
  • isolated does not necessarily require that the protein, polypeptide, peptide or oligopeptide so described has been physically removed from its native environment.
  • polypeptide fragment refers to a polypeptide that has an amino-terminal and/or carboxy-terminal deletion compared to a full-length polypeptide.
  • the polypeptide fragment is a contiguous sequence in which the amino acid sequence of the fragment is identical to the corresponding positions in the naturally-occurring sequence. Fragments typically are at least 5, 6, 7, 8, 9 or 10 amino acids long, preferably at least 12, 14, 16 or 18 amino acids long, more preferably at least 20 amino acids long, more preferably at least 25, 30, 35, 40 or 45, amino acids, even more preferably at least 50 or 60 amino acids long, and even more preferably at least 70 amino acids long.
  • a “modified derivative” refers to polypeptides or fragments thereof that are substantially homologous in primary structural sequence but which include, e.g., in vivo or in vitro chemical and biochemical modifications or which incorporate amino acids that are not found in the native polypeptide. Such modifications include, for example, acetylation, carboxylation, phosphorylation, glycosylation, ubiquitination, labeling, e.g., with radionuclides, and various enzymatic modifications, as will be readily appreciated by those well skilled in the art.
  • a variety of methods for labeling polypeptides and of substituents or labels useful for such purposes are well-known in the art, and include radioactive isotopes such as 125 I, 32 P, 35 S, and 3 H, ligands which bind to labeled antiligands (e.g., antibodies), fluorophores, chemiluminescent agents, enzymes, and antiligands which can serve as specific binding pair members for a labeled ligand.
  • the choice of label depends on the sensitivity required, ease of conjugation with the primer, stability requirements, and available instrumentation.
  • Methods for labeling polypeptides are well-known in the art. See Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates (1992, and Supplements to 2002), hereby incorporated by reference.
  • a “polypeptide mutant” or “mutein” refers to a polypeptide whose sequence contains an insertion, duplication, deletion, rearrangement or substitution of one or more amino acids compared to the amino acid sequence of a native or wild-type protein.
  • a mutein may have one or more amino acid point substitutions, in which a single amino acid at a position has been changed to another amino acid, one or more insertions and/or deletions, in which one or more amino acids are inserted or deleted, respectively, in the sequence of the naturally-occurring protein, and/or truncations of the amino acid sequence at either or both the amino or carboxy termini.
  • a mutein may have the same but preferably has a different biological activity compared to the naturally-occurring protein.
  • a mutein has at least 70% overall sequence homology to its wild-type counterpart. Even more preferred are muteins having 80%, 85% or 90% overall sequence homology to the wild-type protein. In an even more preferred embodiment, a mutein exhibits 95% sequence identity, even more preferably 97%, even more preferably 98% and even more preferably 99% overall sequence identity. Sequence homology may be measured by any common sequence analysis algorithm, such as Gap or Bestfit.
  • Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinity or enzymatic activity, and (5) confer or modify other physicochemical or functional properties of such analogs.
  • Examples of unconventional amino acids include: 4-hydroxyproline, ⁇ -carboxyglutamate, ⁇ -N,N,N-trimethyllysine, ⁇ -N-acetyllysine, O-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, N-methylarginine, and other similar amino acids and imino acids (e.g., 4-hydroxyproline).
  • the left-hand direction is the amino terminal direction and the right hand direction is the carboxy-terminal direction, in accordance with standard usage and convention.
  • a protein has “homology” or is “homologous” to a second protein if the nucleic acid sequence that encodes the protein has a similar sequence to the nucleic acid sequence that encodes the second protein.
  • a protein has homology to a second protein if the two proteins have “similar” amino acid sequences.
  • the term “homologous proteins” is defined to mean that the two proteins have similar amino acid sequences).
  • a homologous protein is one that exhibits 60% sequence homology to the wild type protein, more preferred is 70% sequence homology. Even more preferred are homologous proteins that exhibit 80%, 85% or 90% sequence homology to the wild type protein.
  • a homologous protein exhibits 95%, 97%, 98% or 99% sequence identity.
  • homology between two regions of amino acid sequence is interpreted as implying similarity in function.
  • a “conservative amino acid substitution” is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity).
  • R group side chain
  • a conservative amino acid substitution will not substantially change the functional properties of a protein.
  • the percent sequence identity or degree of homology may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well known to those of skill in the art (see, e.g., Pearson (1990) Methods Enzymol. 183:63-98).
  • the following six groups each contain amino acids that are conservative substitutions for one another: 1) Serine (S), Threonine (T); 2) Aspartic Acid (D), Glutamic Acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Alanine (A), Valine (V), and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W).
  • Sequence homology for polypeptides is typically measured using sequence analysis software.
  • sequence analysis software See, e.g., the Sequence Analysis Software Package of the Genetics Computer Group (GCG), University of Wisconsin Biotechnology Center, 910 University Avenue, Madison, Wis. 53705.
  • GCG Genetics Computer Group
  • Protein analysis software matches similar sequences using measure of homology assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions.
  • GCG contains programs such as “Gap” and “Bestfit” which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1.
  • a preferred algorithm when comparing a inhibitory molecule sequence to a database containing a large number of sequences from different organisms is the computer program BLAST (Altschul et al. (1990) J. Mol. Biol. 215:403-410; Gish and States (1993) Nature Genet. 3:266-272; Madden et al. (1996) Meth. Enzymol. 266:131-141; Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402; Zhang and Madden (1997) Genome Res. 7:649-656), especially blastp or tblastn (Altschul et al., 1997).
  • Preferred parameters for BLASTp are: Expectation value: 10 (default); Filter: seg (default); Cost to open a gap: 11 (default); Cost to extend a gap: 1 (default); Max. alignments: 100 (default); Word size: 11 (default); No. of descriptions: 100 (default); Penalty Matrix: BLOWSUM62.
  • polypeptide sequences compared for homology will generally be at least about 16 amino acid residues, usually at least about 20 residues, more usually at least about 24 residues, typically at least about 28 residues, and preferably more than about 35 residues.
  • database searching using amino acid sequences can be measured by algorithms other than blastp known in the art.
  • polypeptide sequences can be compared using FASTA, a program in GCG Version 6.1.
  • FASTA provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (see Pearson (1990) Methods Enzymol. 183:63-98).
  • percent sequence identity between amino acid sequences can be determined using FASTA with its default parameters (a word size of 2 and the PAM250 scoring matrix), as provided in GCG Version 6.1, herein incorporated by reference.
  • fusion protein refers to a polypeptide comprising a polypeptide or fragment coupled to heterologous amino acid sequences. Fusion proteins are useful because they can be constructed to contain two or more desired functional elements from two or more different proteins.
  • a fusion protein comprises at least 10 contiguous amino acids from a polypeptide of interest, more preferably at least 20 or 30 amino acids, even more preferably at least 40, 50 or 60 amino acids, yet more preferably at least 75, 100 or 125 amino acids.
  • Fusion proteins can be produced recombinantly by constructing a nucleic acid sequence which encodes the polypeptide or a fragment thereof in-frame with a nucleic acid sequence encoding a different protein or peptide and then expressing the fusion protein.
  • a fusion protein can be produced chemically by crosslinking the polypeptide or a fragment thereof to another protein.
  • region refers to a physically contiguous portion of the primary structure of a biomolecule. In the case of proteins, a region is defined by a contiguous portion of the amino acid sequence of that protein.
  • domain refers to a structure of a biomolecule that contributes to a known or suspected function of the biomolecule. Domains may be co-extensive with regions or portions thereof; domains may also include distinct, non-contiguous regions of a biomolecule. Examples of protein domains include, but are not limited to, an Ig domain, an extracellular domain, a transmembrane domain, and a cytoplasmic domain.
  • molecule means any compound, including, but not limited to, a small molecule, peptide, protein, sugar, nucleotide, nucleic acid, lipid, etc., and such a compound can be natural or synthetic.
  • the invention provides methods for producing a glycoprotein having human-like glycosylation in a non-human eukaryotic host cell.
  • a eukaryotic host cell that does not naturally express, or which is engineered not to express, one or more enzymes involved in production of high mannose structures is selected as a starting host cell.
  • Such a selected host cell is engineered to express one or more enzymes or other factors required to produce human-like glycoproteins.
  • a desired host strain can be engineered one enzyme or more than one enzyme at a time.
  • a nucleic acid molecule encoding one or more enzymes or activities may be used to engineer a host strain of the invention.
  • a library of nucleic acid molecules encoding potentially useful enzymes e.g., chimeric enzymes comprising a catalytically active enzyme fragment ligated in-frame to a heterologous subcellular targeting sequence
  • potentially useful enzymes e.g., chimeric enzymes comprising a catalytically active enzyme fragment ligated in-frame to a heterologous subcellular targeting sequence
  • a strain having one or more enzymes with optimal activities or producing the most “human-like” glycoproteins may be selected by transforming target host cells with one or more members of the library.
  • the methods described herein enable one to obtain, in vivo, Man 5 GlcNAc 2 structures in high yield, at least transiently, for the purpose of further modifying it to yield complex N-glycans.
  • a successful scheme to obtain suitable Man 5 GlcNAc 2 structures in appropriate yields in a host cell, such as a lower eukaryotic organism generally involves two parallel approaches: (1) reducing high mannose structures made by endogenous mannosyltransferase activities, if any, and (2) removing 1,2- ⁇ -mannose by mannosidases to yield high levels of suitable Man 5 GlcNAc 2 structures which may be further reacted inside the host cell to form complex, human-like glycoforms.
  • a first step involves the selection or creation of a eukaryotic host cell, e.g., a lower eukaryote, capable of producing a specific precursor structure of Man 5 GlcNAc 2 that is able to accept in vivo GlcNAc by the action of a GlcNAc transferase I (“GnTI”).
  • GnTI GlcNAc transferase I
  • the method involves making or using a non-human eukaryotic host cell depleted in a 1,6 mannosyltransferase activity with respect to the N-glycan on a glycoprotein.
  • the host cell is depleted in an initiating 1,6 mannosyltransferase activity (see below).
  • Such a host cell will lack one or more enzymes involved in the production of high mannose structures which are undesirable for producing human-like glycoproteins.
  • Man 5 GlcNAc 2 is necessary for complex N-glycan formation: Man 5 GlcNAc 2 must be formed in vivo in a high yield (e.g., in excess of 30%), at least transiently, as subsequent mammalian- and human-like glycosylation reactions require Man 5 GlcNAc 2 or a derivative thereof.
  • This step also requires the formation of a particular isomeric structure of Man 5 GlcNAc 2 within the cell at a high yield. While Man 5 GlcNAc 2 structures are necessary for complex N-glycan formation, their presence is by no means sufficient. That is because Man 5 GlcNAc 2 may occur in different isomeric forms, which may or may not serve as a substrate for GlcNAc transferase I. As most glycosylation reactions are not complete, a particular glycosylated protein generally contains a range of different carbohydrate structures (i.e., glycoforms) on its surface.
  • Man 5 GlcNAc 2 produced by the selected host cell must be a productive substrate for enzyme activities along a mammalian glycosylation pathway, e.g., can serve as a substrate for a GlcNAc transferase I activity in vivo, thereby forming the human-like N-glycan intermediate GlcNAcMan 5 GlcNAc 2 in the host cell.
  • at least 10%, more preferably at least 30% and most preferably 50% or more of the Man 5 GlcNAc 2 intermediate produced in the host cell of the invention is a productive substrate for GnTI in vivo.
  • host cells from nature, e.g., existing fungi or other lower eukaryotes that produce significant levels of Man 5 GlcNAc 2 in vivo. As yet, however, no lower eukaryote has been shown to provide such structures in vivo in excess of 1.8% of the total N-glycans (see e.g. Maras et al. (1997) Eur. J. Biochem. 249:701-707). Alternatively, such host cells may be genetically engineered to produce the Man 5 GlcNAc 2 structure in vivo. Methods such as those described in U.S. Pat. No. 5,595,900 may be used to identify the absence or presence of particular glycosyltransferases, mannosidases and sugar nucleotide transporters in a target host cell or organism of interest.
  • the methods of the invention are directed to making host cells which produce glycoproteins having altered, and preferably human-like, N-glycan structures.
  • the methods are directed to making host cells in which oligosaccharide precursors are enriched in Man 5 GlcNAc 2 .
  • a eukaryotic host cell is used that does not express one or more enzymes involved in the production of high mannose structures.
  • Such a host cell may be found in nature or may be engineered, e.g., starting with or derived from one of many such mutants already described in yeasts. Thus, depending on the selected host cell, one or a number of genes that encode enzymes known to be characteristic of non-human glycosylation reactions will have to be deleted.
  • genes and their corresponding proteins have been extensively characterized in a number of lower eukaryotes (e.g., S. cerevisiae, T. reesei, A. nidulans, etc.), thereby providing a list of known glycosyltransferases in lower eukaryotes, their activities and their respective genetic sequence.
  • These genes are likely to be selected from the group of mannosyltransferases, e.g. 1,3 mannosyltransferases (e.g. MNN1 in S. cerevisiae ) (Graham, 1991), 1,2 mannosyltransferases (e.g. KTR/KRE family from S.
  • Preferred lower eukaryotic host cells of the invention are hypermannosylation-minus (och1) mutants of Pichia pastoris or K. lactis.
  • och1 hypermannosylation-minus
  • P. pastoris processes Man 9 GlcNAc 2 structures in the ER with an ⁇ -1,2-mannosidase to yield Man 8 GlcNAc 2 ( FIG. 1A ).
  • Man >9 GlcNAc 2 also known as mannans
  • pastoris is able to add non-terminal phosphate groups, through the action of mannosylphosphate transferases, to the carbohydrate structure. This differs from the reactions performed in mammalian cells, which involve the removal rather than addition of mannose sugars ( FIG. 35A ). It is of particular importance to eliminate the ability of the eukaryotic host cell, e.g., fungus, to hypermannosylate an existing Man 8 GlcNAc 2 structure. This can be achieved by either selecting for a host cell that does not hypermannosylate or by genetically engineering such a cell.
  • genes that are involved in the hypermannosylation process have been identified, e.g., in Pichia pastoris, and by creating mutations in these genes, one can reduce the production of “undesirable” glycoforms.
  • Such genes can be identified by homology to existing mannosyltransferases or their regulators (e.g., OCH1, MNN4, MNN6, MNN1) found in other lower eukaryotes such as C. albicans, Pichia angusta or S. cerevisiae or by mutagenizing the host strain and selecting for a glycosylation phenotype with reduced mannosylation.
  • PCR primers examples of which are shown in Table 2
  • genes or gene fragments encoding such enzymes as probes to identify homologs in DNA libraries of the target or a related organism.
  • TABLE 2 PCR Primers Target Gene(s) PCR primer A PCR primer B in P. pastoris Homologs ATGGCGAAGGCA TTAGTCCTTCCAA 1,6- OCH1 S.
  • GATGGCAGT CTTCCTTC mannosyl- Pichia albicans (SEQ ID NO:3) (SEQ ID NO:4) transferase TAYTGGMGNGTN GCRTCNCCCCANC 1,2 KTR/KRE family, GARCYNGAYATH KYTCRTA mannosyl- S. cerevisiae AA (SEQ ID NO:6) transferases (SEQ ID NO:5)
  • M A or C
  • R A or G
  • W A or T
  • S C or G
  • Y C or T
  • K G or T
  • V A or C or G
  • H A or C or T
  • D A or G or T
  • B C or G or T
  • N G or A or T or C.
  • OCH1 mutants of S. cerevisiae are temperature sensitive and are slow growers at elevated temperatures.
  • Mutants of S. cerevisiae are available, e.g., from Stanford University, and are commercially available from ResGen, Invitrogen Corp. (Carlsbad, Calif.). Mutants that display a normal growth phenotype at elevated temperature, after having been transformed with a P.
  • Such a library can be created by partially digesting chromosomal DNA of P. pastoris with a suitable restriction enzyme and, after inactivating the restriction enzyme, ligating the digested DNA into a suitable vector, which has been digested with a compatible restriction enzyme.
  • Suitable vectors include, e.g., pRS314, a low copy (CEN6/ARS4) plasmid based on pBluescript containing the Trp1 marker (Sikorski and Hieter (1989) Genetics 122:19-27) and pFL44S, a high copy (2 ⁇ ) plasmid based on a modified pUC 19 containing the URA3 marker (Bonneaud et al. (1991) Yeast 7:609-615).
  • Such vectors are commonly used by academic researchers and similar vectors are available from a number of different vendors (e.g., Invitrogen (Carlsbad, Calif.); Pharmacia (Piscataway, N.J.); New England Biolabs (Beverly, Mass.)). Further examples include pYES/GS, 2 ⁇ origin of replication based yeast expression plasmid from Invitrogen, or Yep24 cloning vehicle from New England Biolabs.
  • the DNA library After ligation of the chromosomal DNA and the vector, one may transform the DNA library into a strain of S. cerevisiae with a specific mutation and select for the correction of the corresponding phenotype. After sub-cloning and sequencing the DNA fragment that is able to restore the wild-type phenotype, one may use this fragment to eliminate the activity of the gene product encoded by OCH1 in P. pastoris using in vivo mutagenesis and/or recombination techniques well-known to those skilled in the art.
  • genomic sequence of a particular host cell e.g., fungus
  • a known 1,6 mannosyltransferase gene e.g., OCH1 from S. cerevisiae
  • Nucleic acid sequence homology alone is not enough to prove, however, that one has identified and isolated a homolog encoding an enzyme having the same activity.
  • Homologs to several S. cerevisiae mannosyltransferases have been identified in P. pastoris using these approaches.
  • Homologous genes often have similar functions to genes involved in the mannosylation of proteins in S. cerevisiae and thus their deletion may be used to manipulate the glycosylation pattern in P. pastoris or, by analogy, in any other host cell, e.g., fungus, plant, insect or animal cells, with similar glycosylation pathways.
  • URA3 may be used as a marker to ensure the selection of a transformants that have integrated a construct. By flanking the URA3 marker with direct repeats one may first select for transformants that have integrated the construct and have thus disrupted the target gene.
  • Eliminating specific mannosyltransferases, such as 1,6 mannosyltransferase (OCH1) or mannosylphosphate transferases (MNN6, or genes complementing lbd mutants) or regulators (MNN4) in P. pastoris enables one to create engineered strains of this organism which synthesize primarily Man 8 GlcNAc 2 and which can be used to further modify the glycosylation pattern to resemble more complex glycoform structures, e.g., those produced in mammalian, e.g., human cells.
  • a preferred embodiment of this method utilizes DNA sequences encoding biochemical glycosylation activities to eliminate similar or identical biochemical functions in P. pastoris to modify the glycosylation structure of glycoproteins produced in the genetically altered P. pastoris strain.
  • Methods used to engineer the glycosylation pathway in yeasts as exemplified herein can be used in filamentous fungi to produce a preferred substrate for subsequent modification.
  • Strategies for modifying glycosylation pathways in A. niger and other filamentous fungi, for example, can be developed using protocols analogous to those described herein for engineering strains to produce human-like glycoproteins in yeast.
  • Undesired gene activities involved in 1,2 mannosyltransferase activity, e.g., KTR/KRE homologs, are modified or eliminated.
  • a filamentous fungus such as Aspergillus, is a preferred host because it lacks the 1,6 mannosyltransferase activity and as such, one would not expect a hypermannosylating gene activity, e.g. OCH1, in this host.
  • other desired activities e.g., ⁇ -1,2-mannosidase, UDP-GlcNAc transporter, glycosyltransferase (GnT), galactosyltransferase (GalT) and sialyltransferase (ST) involved in glycosylation are introduced into the host using the targeting methods of the invention.
  • the method of the invention involves making or using a host cell which is diminished or depleted in the activity of an initiating ⁇ -1,6-mannosyltransferase, i.e., an initiation specific enzyme that initiates outer chain mannosylation on the ⁇ -1,3 arm of the Man 3 GlcNAc 2 core structure.
  • an initiating ⁇ -1,6-mannosyltransferase i.e., an initiation specific enzyme that initiates outer chain mannosylation on the ⁇ -1,3 arm of the Man 3 GlcNAc 2 core structure.
  • this enzyme is encoded by the OCH1 gene.
  • Disruption of the OCH1 gene in S. cerevisiae results in a phenotype in which N-linked sugars completely lack the poly-mannose outer chain.
  • host cells selected or produced according to this invention which accumulate oligosaccharides having seven or fewer mannose residues may produce hypoglycosylated N-glycans that will likely be poor substrates for Och1p (see, e.g., Nakayama et al. (1 997) FEBS Lett. 412(3):547-50).
  • the OCH1 gene was cloned from P. pastoris (Example 1) and K. lactis (Example 9), as described.
  • the nucleic acid and amino acid sequences of the OCH1 gene from K. lactis are set forth in SEQ ID NOs:7 and 8.
  • a construct was made from each clone to delete the OCH1 gene from the genome of P. pastoris and K. lactis (Examples 1 and 9, respectively).
  • Host cells depleted in initiating ⁇ -1,6-mannosyltransferase activity and engineered to produce N-glycans having a Man 5 GlcNAc 2 carbohydrate structure were thereby obtained (see, e.g., FIGS. 5, 6 , and 12 ; Examples 4 and 9).
  • the invention provides an isolated nucleic acid molecule having a nucleic acid sequence comprising or consisting of at least forty-five, preferably at least 50, more preferably at least 60 and most preferably 75 or more nucleotide residues of the K. lactis OCH1 gene (SEQ ID NO: 7), and homologs, variants and derivatives thereof.
  • the invention also provides nucleic acid molecules that hybridize under stringent conditions to the above-described nucleic acid molecules.
  • isolated polypeptides including muteins, allelic variants, fragments, derivatives, and analogs
  • vectors including expression vectors, which comprise the above nucleic acid molecules of the invention, as described further herein.
  • host cells transformed with the nucleic acid molecules or vectors of the invention are provided.
  • the invention further provides methods of making or using a non-human eukaryotic host cell diminished or depleted in an alg gene activity (i.e., alg activities, including equivalent enzymatic activities in non-fungal host cells) and introducing into the host cell at least one glycosidase activity.
  • the glycosidase activity is introduced by causing expression of one or more mannosidase activities within the host cell, for example, by activation of a mannosidase activity, or by expression from a nucleic acid molecule of a mannosidase activity, in the host cell.
  • the method involves making or using a host cell diminished or depleted in the activity of one or more enzymes that transfer a sugar residue to the 1,6 arm of lipid-linked oligosaccharide precursors ( FIG. 13 ).
  • a host cell of the invention is selected for or is engineered by introducing a mutation in one or more of the genes encoding an enzyme that transfers a sugar residue (e.g., mannosylates) the 1,6 arm of a lipid-linked oligosaccharide precursor.
  • the sugar residue is more preferably mannose, is preferably a glucose, GlcNAc, galactose, sialic acid, fucose or GlcNAc phosphate residue.
  • the activity of one or more enzymes that mannosylate the 1,6 arm of lipid-linked oligosaccharide precursors is diminished or depleted.
  • the method may further comprise the step of introducing into the host cell at least one glycosidase activity (see below).
  • the invention provides a method for producing a human-like glycoprotein in a non-human host, wherein the glycoprotein comprises an N-glycan having at least two GlcNAcs attached to a trimannose core structure.
  • the method is directed to making a host cell in which the lipid-linked oligosaccharide precursors are enriched in Man X GlcNAc 2 structures, where X is 3, 4 or 5 ( FIG. 14 ).
  • These structures are transferred in the ER of the host cell onto nascent polypeptide chains by an oligosaccharyl-transferase and may then be processed by treatment with glycosidases (e.g., ⁇ -mannosidases) and glycosyltransferases (e.g., GnTI) to produce N-glycans having GlcNAcMan X GlcNAc 2 core structures, wherein X is 3, 4 or 5, and is preferably 3 ( FIGS.
  • glycosidases e.g., ⁇ -mannosidases
  • GnTI glycosyltransferases
  • N-glycans having a GlcNAcMan X GlcNAc 2 core structure where X is greater than 3 may be converted to GlcNAcMan 3 GlcNAc 2 , e.g., by treatment with an ⁇ -1,3 and/or ⁇ -1,2-1,3 mannosidase activity, where applicable.
  • GlcNAcMan 3 GlcNAc 2 by treatment with glycosyltransferases (e.g., GnTII) produces GlcNAc 2 Man 3 GlcNAc 2 core structures which may then be modified, as desired, e.g., by ex vivo treatment or by heterologous expression in the host cell of a set of glycosylation enzymes, including glycosyltransferases, sugar transporters and mannosidases (see below), to become human-like N-glycans.
  • glycosyltransferases e.g., GnTII
  • Preferred human-like glycoproteins which may be produced according to the invention include those which comprise N-glycans having seven or fewer, or three or fewer, mannose residues; comprise one or more sugars selected from the group consisting of galactose, GlcNAc, sialic acid, and fucose; and comprise at least one oligosaccharide branch comprising the structure NeuNAc-Gal-GlcNAc-Man.
  • the host cell has diminished or depleted Do1-P-Man:Man 5 GlcNAc 2 -PP-Do1 Mannosyltransferase activity, which is an activity involved in the first mannosylation step from Man 5 GlcNAc 2 -PP-Do1 to Man 6 GlcNAc 2 -PP-Do1 at the luminal side of the ER (e.g., ALG3 FIG. 13 ; FIG. 14 ).
  • this enzyme is encoded by the ALG3 gene. As described above, S.
  • cerevisiae cells harboring a leaky alg3-1 mutation accumulate Man 5 GlcNAc 2 -PP-Do1 and cells having a deletion in alg3 appear to transfer Man 5 GlcNAc 2 structures onto nascent polypeptide chains within the ER.
  • host cells will accumulate N-glycans enriched in Man 5 GlcNAc 2 structures which can then be converted to GlcNAc 2 Man 3 GlcNAc 2 by treatment with glycosidases (e.g., with ⁇ -1,2 mannosidase, ⁇ -1,3 mannosidase, or ⁇ -1,2-1,3 mannosidase activities) and glycosyltransferase activites (e.g., GnTI, GnTII) ( FIG. 14 ; FIG. 35B ).
  • glycosidases e.g., with ⁇ -1,2 mannosidase, ⁇ -1,3 mannosidase, or ⁇ -1,2-1,3 mannosidase activities
  • glycosyltransferase activites e.g., GnTI, GnTII
  • degenerate primers were designed based on an alignment of Alg3 protein sequences from S. cerevisiae, D. melanogaster and humans ( H. sapiens ) ( FIGS. 16 and 17 ), and were used to amplify a product from P. pastoris genomic DNA.
  • the resulting PCR product was used as a probe to identify and isolate a P. pastoris genomic clone comprising an open reading frame (ORF) that encodes a protein having 35% overall sequence identity and 53% sequence similarity to the S. cerevisiae ALG3 gene ( FIGS. 18 and 19 ).
  • This P. pastoris gene is referred to herein as “PpALG3”.
  • the ALG3 gene was similarly identified and isolated from K. lactis (Example 10; FIGS. 20 and 21 ).
  • the invention provides an isolated nucleic acid molecule having a nucleic acid sequence comprising or consisting of at least forty-five, preferably at least 50, more preferably at least 60 and most preferably 75 or more nucleotide residues of the P. pastoris ALG3 gene ( FIG. 18 ) and the K. lactis ALG3 gene ( FIG. 20 ), and homologs, variants and derivatives thereof.
  • the invention also provides nucleic acid molecules that hybridize under stringent conditions to the above-described nucleic acid molecules.
  • isolated polypeptides including muteins, allelic variants, fragments, derivatives, and analogs
  • P. pastoris and K. lactis ALG3 gene products are shown in FIGS. 18 and 20 ).
  • vectors including expression vectors, which comprise a nucleic acid molecule of the invention, as described further herein.
  • a construct was made to delete the PpALG3 gene from the genome of P. pastoris (Example 10).
  • This strain was used to generate a host cell depleted in Do1-P-Man:Man 5 GlcNAc 2 -PP-Do1 Mannosyltransferase activity and produce lipid-linked Man 5 GlcNAc 2 -PP-Do1 precursors which are transferred onto nascent polypeptide chains to produce N-glycans having a Man 5 GlcNAc 2 carbohydrate structure.
  • such a host cell may be engineered by expression of appropriate mannosidases to produce N-glycans having the desired Man 3 GlcNAc 2 core carbohydrate structure.
  • Expression of GnTs in the host cell enables the modified host cell to produce N-glycans having one or two GlcNAc structures attached to each arm of the Man3 core structure (i.e., GlcNAc 1 Man 3 GlcNAc 2 , GlcNAc 2 Man 3 GlcNAc 2 , or GlcNAc 3 Man 3 GlcNAc 2 ; see FIG. 15 ).
  • These structures may be processed further using the methods of the invention to produce human-like N-glycans on proteins which enter the secretion pathway of the host cell.
  • the method of the invention involves making or using a host cell which is both (a) diminished or depleted in the activity of an alg gene or in one or more activities that mannosylate N-glycans on the ⁇ -1,6 arm of the Man 3 GlcNAc 2 (“Man3”) core carbohydrate structure; and (b) diminished or depleted in the activity of an initiating ⁇ -1,6-mannosyltransferase, i.e., an initiation specific enzyme that initiates outer chain mannosylation (on the ⁇ -1,3 arm of the Man3 core structure).
  • this enzyme is encoded by the OCH1 gene. Disruption of the och1 gene in S.
  • the host cells selected or produced according to this invention which accumulate lipid-linked oligosaccharides having seven or fewer mannose residues will, after transfer, produce hypoglycosylated N-glycans that will likely be poor substrates for Och1p (see, e.g., Nakayama et al. (1997) FEBS Lett. 412(3):547-50).
  • the invention additionally provides a method for producing a human-like glycoprotein in a lower eukaryotic host cell by expressing an N-acetylglucosaminyltransferase III activity (including a full-length enzyme, homologs, variants, derivatives, and catalytically active fragments thereof).
  • a host cell e.g., P. pastoris
  • a host cell is engineered to produce more human-like N-glycans, e.g., by activation of an N-acetylglucosaminyltransferase III activity or by expression from a nucleic acid molecule of an N-acetylglucosaminyltransferase III activity.
  • gene-specific primers are designed to complement the homologous regions of a GnTIII gene, preferably a mammalian GnTIII gene (e.g., mouse GnTIII) ( FIG. 24 ), sequences for which are readily available in the art (e.g., Genbank Accession No. L39373) and are PCR amplified.
  • a mammalian GnTIII gene e.g., mouse GnTIII
  • sequences for which are readily available in the art e.g., Genbank Accession No. L39373
  • the invention provides a method for producing a human-like glycoprotein in a lower eukaryote (e.g., P. pastoris ), wherein the glycoprotein comprises an N-glycan exhibiting a bisecting GlcNAc on a trimannose or trimannosyl (Man 3 GlcNAc 2 ) core structure.
  • a lower eukaryote e.g., P. pastoris
  • the glycoprotein comprises an N-glycan exhibiting a bisecting GlcNAc on a trimannose or trimannosyl (Man 3 GlcNAc 2 ) core structure.
  • GlcNAcMan 3 GlcNAc 2 (which may be produced by reacting a trimannose core with N-acetylglucosaminyltransferase I (“GnTI”) activity, but which is typically produced by trimming of GlcNAcMan 5 GlcNAc 2 by an ⁇ -1,3/ ⁇ -1,6-mannosidase activity, such as Mannosidase II (Hamilton et al. (2003) Science 301:1244-46)) is reacted with an N-acetylglucosaminyltransferase III activity to produce a bisected GlcNAc 2 Man 3 GlcNAc 2 .
  • GnTI N-acetylglucosaminyltransferase I
  • the invention provides GnTIII activity, which transfers ⁇ -1,4 GlcNAc onto substrates that are capable of accepting the bisecting GlcNAc in lower eukaryotes.
  • the invention provides a method for producing a human-like glycoprotein in a lower eukaryote (e.g., P. pastoris ), wherein the glycoprotein comprises an N-glycan exhibiting a bisecting GlcNAc on a trimannose or trimannosyl (Man 3 GlcNAc 2 ) core structure having at least two GlcNAcs attached to the trimannose core.
  • a lower eukaryote e.g., P. pastoris
  • the glycoprotein comprises an N-glycan exhibiting a bisecting GlcNAc on a trimannose or trimannosyl (Man 3 GlcNAc 2 ) core structure having at least two GlcNAcs attached to the trimannose core.
  • Man 3 GlcNAc 2 is reacted with a GnTI activity and then with an N-acetylglucosaminyltransferase II (“GnTII”) activity and a GnTIII activity (in either order) to produce a bisected GlcNAc 3 Man 3 GlcNAc 2 ( FIG. 38 ).
  • GnTII N-acetylglucosaminyltransferase II
  • GnTIII activity in either order
  • the bisected trimannosyl core structure of this embodiment may also contain an additional mannosyl group in place of a GlcNAc residue.
  • GlcNAcMan 4 GlcNAc 2 may be reacted with a GnTIII activity to produce a bisected GlcNAc 2 Man 4 GlcNAc 2 .
  • the invention also provides a method for producing a more human-like glycoprotein in a lower eukaryote (e.g. P. pastoris ), wherein the glycoprotein produced comprises an N-glycan having at least two GlcNAcs attached to a pentamannose core structure (Man 5 GlcNAc 2 ) and which exhibits a bisected N-glycan.
  • a pentamannose core structure Man 5 GlcNAc 2
  • GnTIII activity to produce a bisected GlcNAcMan 5 GlcNAc 2 and GlcNAc 2 Man 5 GlcNAc 2 structure.
  • a pentamannose core structure produced via the mutation of och1 and alg3 genes is reacted with ⁇ 1,2-mannosidase, GnTI, GnTII and GnTIII activities and UDP-GlcNAc to produce a bisected GlcNAc 3 Man 3 GlcNAc 2 glycan ( FIG. 35B ).
  • a pentamannose core structure is reacted with GnTI and GnTIII activities (in either order or in combination) to produce a bisected GlcNAc 2 Man 5 GlcNAc 2 structure ( FIG. 37 ).
  • a pVA53 construct comprising the S. cerevisiae MNN2(s) leader (GenBank Accession No. NP — 009571) fused to a catalytically active GnTIII domain from mouse (GnTIII ⁇ 32) is expressed in a P. pastoris strain YSH-1 (Example 13) thereby producing N-glycans having a bisected GlcNAc 2 Man 5 GlcNAc 2 structure (Example 20).
  • FIG. 26 (bottom) displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in the above-mentioned strain, which is designated PBP26 ( FIG. 36 ), exhibiting a predominant peak at 1666 m/z [a], which corresponds to bisected GlcNAc 2 Man 5 GlcNAc 2 .
  • FIG. 26 (top) displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in strain YSH-1 lacking the pVA53 construct.
  • a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 50 mole % of a GlcNAc 2 Man 5 GlcNAc 2 or at least 50 mole % of a GlcNAc 2 Man 3 GlcNAc 2 structure having a bisecting GlcNAc.
  • the mole percent of the glycans is in reference to percent of total neutral glycans as detected by MALDI-TOF.
  • GlcNAc 2 Man 3 GlcNAc 2 having a bisecting GlcNAc is produced at 20% and GlcNAc 3 Man 3 GlcNAc 2 is produced at 25% on a target protein
  • the total amount of transiently produced GlcNAc 2 Man 3 GlcNAc 2 having a bisecting GlcNAc is 45%, because GlcNAc 3 Man 3 GlcNAc 2 is a product of a GlcNAc 2 Man 3 GlcNAc 2 having a bisecting GlcNAc further reacted with GnTII.
  • a pVA55 construct comprising the S. cerevisiae MNN2(1) leader (GenBank Accession No. NP — 009571) fused to a catalytically active GnTIII domain from mouse (GnTIII A32) is expressed in a P. pastoris strain (YSH-1) thereby producing N-glycans GlcNAcMan 5 GlcNAc 2 and bisected N-glycans GlcNAc 2 Man 5 GlcNAc 2 structure. As shown in FIG. 27 (bottom), these structures correspond to peaks at 1463 m/z and 1667 m/z, respectively. (For comparison, FIG.
  • a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 20 mole % of a GlcNAc 2 Man 5 GlcNAc 2 or at least 20 mole % of a GlcNAc 2 Man 3 GlcNAc 2 structure having a bisecting GlcNAc.
  • a pVA53 construct comprising the S. cerevisiae MNN2(s) leader (GenBank Accession No. NP — 009571) fused to a catalytically active GnTIII domain from mouse (GnTIII ⁇ 32) is expressed in a P. pastoris strain YSH-44 (Example 15) thereby producing N-glycans having a bisected GlcNAc 3 Man 3 GlcNAc 2 structure (Example 20).
  • FIG. 30 displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in the above-mentioned strain designated as YSH-57, exhibiting a predominant peak at 1542 m/z [y], which corresponds to the bisected glycan GlcNAc 3 Man 3 GlcNAc 2 .
  • FIG. 29 displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in strain YSH-44 lacking the pVA53 construct. The predominant peak at 1356 m/z [x] in FIG.
  • a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 80 mole % of a GlcNAc 3 Man 3 GlcNAc 2 structure having a bisecting GlcNAc.
  • the mole percent of the glycans is in reference to percent of total neutral glycans as detected by MALDI-TOF.
  • a pVA53 construct comprising the S. cerevisiae MNN2(s) leader (GenBank Accession No. NP — 009571) fused to a catalytically active GnTIII domain from mouse (GnTIII ⁇ 32) is expressed in a P. pastoris strain (PBP6-5) (Example 11) thereby producing N-glycans having a GlcNAc 2 Man 3 GlcNAc 2 and a bisected GlcNAc 3 Man 3 GlcNAc 2 structure. As shown in FIG. 32 , these structures correspond to peaks at 1340 m/z and 1543 m/z, respectively.
  • a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 20 mole % of a GlcNAc 3 Man 3 GlcNAc 2 structure having a bisecting GlcNAc in an alg3 mutant host cell.
  • the invention provides methods for producing a human-like glycoprotein in a lower eukaryote, wherein the glycoprotein comprises a Man 5 GlcNAc 2 core structure or a Man 3 GlcNAc 2 core structure, and wherein the core structure is further modified by two or more GlcNAcs.
  • the core structure is further modified by two or more GlcNAcs.
  • 10% or more of the core structures are modified by the two or more GlcNAcs.
  • 20%, 30%, 40%, 50%, 60%, 70%, 80% or even more of the core structures are so modified.
  • one of the GlcNAcs is a bisecting GlcNAc.
  • a combinatorial nucleic acid library which encodes at least one GnTIII catalytic domain is used to express a GnTIII activity in a lower eukaryotic host cell (Example 18).
  • a library of the invention comprises a sublibrary of leader sequences fused in frame to a single nucleic acid molecule or a sublibrary of nucleic acid molecules comprising GnTIII sequences, one or more of which encode a catalytic domain having GnTIII activity in the host cell.
  • a single nucleic acid molecule or a sublibrary of nucleic acid molecules comprising leader sequences is fused in frame to a sublibrary of nucleic acid molecules comprising GnTIII sequences, one or more of which encode a catalytic domain having GnTIII activity in the host cell.
  • GnTIII sequences one or more of which encode a catalytic domain having GnTIII activity in the host cell.
  • the proper targeting of an enzyme responsible for a given step in the glycosylation pathway to the appropriate subcellular location and the sufficiency of the enzyme's activity at the particular pH of that subcellular location are important factors in the production of glycoproteins having N-glycans with the desired structures.
  • the use of combinatorial libraries of fusion proteins to generate diverse populations of enzyme chimeras and the screening of these libraries in transformed cells provides a powerful method to identify host strains with the activity of interest in the appropriate location.
  • the enzyme activity is located such that an N-glycan-containing glycoprotein expressed in the cell is capable of reacting with the activity during the secretion process.
  • FIG. 28 shows a pVB51 construct comprising the K. lactis GNT(s) leader (GenBank Accession No. AF106080) fused to a catalytically active GnTIII domain from mouse (GnTIII A32) expressed in a P. pastoris strain YSH-1, which does not readily exhibit GnTIII activity. (For comparison, FIG.
  • FIG. 28 (top) displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in strain YSH-1 lacking the pVA53 construct.
  • the predominant peak corresponds to unmodified GlcNAcMan 5 GlcNAc 2 at 1461 m/z.
  • the predominant peak in FIG. 28 (bottom) at 1463 m/z, which correlates to the mass of GlcNAcMan 5 GlcNAc 2 is observed.
  • a second peak at 1726 m/z which does not correlate to the mass of GlcNAc 2 Man 5 GlcNAc 2 is also observed. It is contemplated that these and other such combinations may be useful, with or without slight modifications using techniques well known in the art, when they are expressed, e.g., in other host cells including those which have been modified to produce human-like glycoforms.
  • Example 6 provides an example of assay conditions useful for measuring extracellular ⁇ -1,2-mannosidase activity.
  • Examples 22 and 23 also provide examples of assays for glycosyltransferase activity (GnTIII) in the medium. See also Table 9, below, and Choi et al. (20033 Proc. Natl. Acad. Sci. U.S.A. 100(9):5022-27.
  • an enzyme activity is substantially intracellular when less than 10% of the enzyme activity is measurable in the extracellular medium.
  • a host cell may be engineered by the expression of appropriate glycosyltransferases (e.g., N-acetylglucosaminyltransferase) to produce N-glycans having the desired carbohydrate structures (e.g., GlcNAc 2 Man 3 GlcNAc 2 , GlcNAc 3 Man 3 GlcNAc 2 ).
  • appropriate glycosyltransferases e.g., N-acetylglucosaminyltransferase
  • GnTs in the host cell e.g., by targeting a nucleic acid molecule or a library of nucleic acid molecules as described below and in Choi et al. (2003) Proc. Natl. Acad. Sci. US.A.
  • co-expression of appropriate UDP-sugar-transporter(s) and transferase(s) will cap the terminal ⁇ -1,6 and ⁇ -1,3 residues as well as the middle mannose with GlcNAc, resulting in the precursor for mammalian-type complex (e.g. GlcNAc 3 Man 3 GlcNAc 2 ) and hybrid N-glycosylation.
  • mammalian-type complex e.g. GlcNAc 3 Man 3 GlcNAc 2
  • These peptide-bound N-linked oligosaccharide chains then serve as a precursor for further modification to a mammalian-type oligosaccharide structure.
  • Subsequent expression of galactosyl-tranferases and genetically engineering the capacity to transfer sialylic acid to the termini will produce a mammalian-type (e.g., human-like) N-glycan structure.
  • a preferred host cell of the invention is a lower eukaryotic cell, e.g., yeast, a unicellular and multicellular or filamentous fungus.
  • yeast e.g., a unicellular and multicellular or filamentous fungus.
  • a wide variety of host cells are envisioned as being useful in the methods of the invention.
  • Plant cells or insect cells may be engineered to express a human-like glycoprotein according to the invention.
  • a variety of non-human, mammalian host cells may be altered to express more human-like or otherwise altered glycoproteins using the methods of the invention.
  • any eukaryotic host cell may be used in conjunction with a library of the invention to express one or more chimeric proteins which is targeted to a subcellular location, e.g., organelle, in the host cell where the activity of the protein is modified, and preferably is enhanced.
  • a protein is preferably—but need not necessarily be—an enzyme involved in protein glycosylation, as exemplified herein. It is envisioned that any protein coding sequence may be targeted and selected for modified activity in a eukaryotic host cell using the methods described herein.
  • Lower eukaryotes that are able to produce glycoproteins having the attached N-glycan Man 5 GlcNAc 2 are particularly useful because (a) lacking a high degree of mannosylation (e.g., greater than 8 mannoses per N-glycan, or especially 30-40 mannoses), they show reduced immunogenicity in humans; and (b) the N-glycan is a substrate for further glycosylation reactions to form an even more human-like glycoform, e.g., by the action of GlcNAc transferase I ( FIG. 1B ; ⁇ 1,2 GnTI) to form GlcNAcMan 5 GlcNAc 2 .
  • GlcNAc transferase I FIG. 1B ; ⁇ 1,2 GnTI
  • a yield is obtained of greater than 30 mole %, more preferably a yield of 50, 60, 70, 80, 90, or even 100 mole %, glycoproteins with N-glycans having a Man 5 GlcNAc 2 structure.
  • more than 50% of the Man 5 GlcNAc 2 structure is shown to be a substrate for a GnTI activity and can serve as such a substrate in vivo.
  • Preferred lower eukaryotes of the invention include but are not limited to: Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reseei, Chrysosporium lucknowense, Fusarium sp. Fusarium gramineum, Fu
  • the method is directed to making a host cell in which the oligosaccharide precursors are enriched in Man 5 GlcNAc 2 .
  • These structures are desirable because they may then be processed by treatment in vitro, for example, using the method of Maras and Contreras, U.S. Pat. No. 5,834,251.
  • precursors enriched in Man 5 GlcNAc 2 are processed by at least one further glycosylation reaction in vivo—with glycosidases (e.g., ⁇ -mannosidases) and glycosyltransferases (e.g., GnTI)—to produce human-like N-glycans.
  • glycosidases e.g., ⁇ -mannosidases
  • GnTI glycosyltransferases
  • Oligosaccharide precursors enriched in Man 5 GlcNAc 2 are preferably processed to those having GlcNAcMan X GlcNAc 2 core structures, wherein X is 3, 4 or 5, and is preferably 3.
  • N-glycans having a GlcNAcMan X GlcNAc 2 core structure where X is greater than 3 may be converted to GlcNAcMan 3 GlcNAc 2 , e.g., by treatment with an ⁇ -1,3 and/or ⁇ -1,6 mannosidase activity, where applicable.
  • GlcNAcMan 3 GlcNAc 2 by treatment with glycosyltransferases (e.g., GnTII) produces GlcNAc 2 Man 3 GlcNAc 2 core structures which may then be modified, as desired, e.g., by ex vivo treatment or by heterologous expression in the host cell of additional glycosylation enzymes, including glycosyltransferases, sugar transporters and mannosidases (see below), to become human-like N-glycans.
  • glycosyltransferases e.g., GnTII
  • Preferred human-like glycoproteins which may be produced according to the invention include those which comprise N-glycans having seven or fewer, or three or fewer, mannose residues; and which comprise one or more sugars selected from the group consisting of galactose, GlcNAc, sialic acid, and fucose.
  • Another preferred non-human host cell of the invention is a lower eukaryotic cell, e.g., a unicellular or filamentous fungus, which is diminished or depleted in the activity of one or more alg gene activities (including an enzymatic activity which is a homolog or equivalent to an alg activity).
  • Another preferred host cell of the invention is diminished or depleted in the activity of one or more enzymes (other than alg activities) that mannosylate the ⁇ -1,6 arm of a lipid-linked oligosaccharide structure.
  • eukaryotic host cells While lower eukaryotic host cells are preferred, a wide variety of host cells having the aforementioned properties are envisioned as being useful in the methods of the invention. Plant cells, for instance, may be engineered to express a human-like glycoprotein according to the invention. Likewise, a variety of non-human, mammalian host cells may be altered to express more human-like glycoproteins using the methods of the invention. An appropriate host cell can be engineered, or one of the many such mutants already described in yeasts may be used.
  • a preferred host cell of the invention, as exemplified herein, is a hypermannosylation-minus (OCH1) mutant in Pichia pastoris which has further been modified to delete the alg3 gene.
  • the invention additionally provides lower eukaryotic host cells capable of producing glycoproteins having bisected N-glycans, such as bisected GlcNAcMan 5 GlcNAc 2 , GlcNAc 2 Man 5 GlcNAc 2 , GlcNAc 2 Man 3 GlcNAc 2 , and, preferably, GlcNAc 3 Man 3 GlcNAc 2 .
  • the host cells comprise a GnTIII activity.
  • the host cells further comprise one or more activities selected from: GnTI, GnTII, GnTIV, and GnTV.
  • Preferred host cells express GnTI, GnTII, and GnTIII.
  • Other preferred host cells additionally express GnTIV and/or GnTV. Even more preferably, the one or more GnT activities of the host cells are substantially intracellular.
  • host cells comprising the one or more GnT activities produce N-glycans comprising structures, including but not limited to GlcNAcMan 3 GlcNAc 2 , GlcNAcMan 4 GlcNAc 2 , or GlcNAcMan 5 GlcNAc 2 , that are capable of reacting with a GnTIII enzyme activity to produce corresponding bisected N-glycans.
  • the enzyme activities thereby convert glycoproteins containing these N-glycans into forms with new and more desirable properties. Because GnTIII is currently understood to inhibit additional GnT activity in mammalian cells, the skilled artisan should appreciate that sequential glycosylation reaction may or may not be of importance.
  • the present invention contemplates, however, the addition of GnTI and GnTIII in either order or together.
  • other enzyme activities within the cell such as, e.g., one or more desired mannosidase activities (e.g., ⁇ 1,2 mannosidase, Mannosidase I, Mannosidase II), may act in concert with the GnT activities to generate yet other human-like glycoproteins of interest (see FIG. 1B ).
  • a mannosidase II or a catalytically active fragment thereof is introduced into the host cell to trim the ⁇ 1,3 and ⁇ 1,6 mannose containing arms of a bisected pentamannose core structure such as GlcNAc 2 Man 5 GlcNAc 2 .
  • the resulting glycans e.g., bisected GlcNAc 2 Man 4 GlcNAc 2 and GlcNAc 2 Man 3 GlcNAc 2
  • the host cells comprise a Man 5 GlcNAc 2 core structure or a Man 3 GlcNAc 2 core structure modified by two or more GlcNAcs. It should be understood that either core structure may include further modifications in addition to the modification by GlcNAc. Preferably, 10% or more of the core structures are modified by GlcNAcs. Most preferably, 20%, 30%, 40%, 50%, 60%, 70%, 80% or even more of the core structures contain the GlcNAc modification.
  • Formation of complex N-glycan synthesis is a sequential process by which specific sugar residues are removed and attached to the core oligosaccharide structure. In higher eukaryotes, this is achieved by having the substrate sequentially exposed to various processing enzymes. These enzymes carry out specific reactions depending on their particular location within the entire processing cascade.
  • This “assembly line” consists of ER, early, medial and late Golgi, and the trans Golgi network all with their specific processing environment.
  • glycosyltransferases e.g., glycosyltransferases, glycosidases, phosphatases and transporters
  • glycosyltransferases e.g., glycosyltransferases, glycosidases, phosphatases and transporters
  • one goal of the methods described herein is to achieve a robust protein production strain that is able to perform well in an industrial fermentation process
  • the integration of multiple genes into the host cell chromosome involves careful planning.
  • one or more genes which encode enzymes known to be characteristic of non-human glycosylation reactions are preferably deleted.
  • the engineered cell strain is transformed with a range of different genes encoding desired activities, and these genes are transformed in a stable fashion, thereby ensuring that the desired activity is maintained throughout the fermentation process.
  • enzyme activities may be engineered singly or multiply into the host using methods of the invention: sialyltransferases, mannosidases, fucosyltransferases, galactosyltransferases, GlcNAc transferases, ER and Golgi specific transporters (e.g. syn- and antiport transporters for UDP-galactose and other precursors), other enzymes involved in the processing of oligosaccharides, and enzymes involved in the synthesis of activated oligosaccharide precursors such as UDP-galactose and CMP-N-acetylneuraminic acid.
  • enzyme activities are introduced on one or more nucleic acid molecules (see also below).
  • Nucleic acid molecules may be introduced singly or multiply, e.g., in the context of a nucleic acid library such as a combinatorial library of the invention. It is to be understood, however, that single or multiple enzymatic activities may be introduced into a host cell in any fashion, including but not limited to protein delivery methods and/or by use of one or more nucleic acid molecules without necessarily using a nucleic acid library or combinatorial library of the invention.
  • nucleic acid molecules encoding GnTI, II, III, IV or V may be inserted into appropriate expression vectors under the transcriptional control of promoters and other expression control sequences capable of driving transcription in a selected host cell of the invention, e.g., a fungal host such as Pichia sp., Kluyveromyces sp.
  • GnT enzymes may be actively expressed in a host cell of choice for production of a human-like complex glycoprotein (e.g., Examples 8, 20, and 21).
  • glycosyltransferases have been cloned and expressed in S. cerevisiae (GalT, GnTI), Aspergillus nidulans (GnTI) and other fungi, without however demonstrating the desired outcome of “humanization” on the glycosylation pattern of the organisms (Yoshida et al. (1999) Glycobiology 9(l):53-8; Kalsner et al. (1995) Glycoconj. J 12(3):360-370). It was speculated that the carbohydrate structure required to accept sugars by the action of such glycosyltransferases was not present in sufficient amounts, which most likely contributed to the lack of complex N-glycan formation.
  • a preferred method of the invention provides the functional expression of a GnT, such as GnTI, GnTII, and GnTIII, in the early, medial or late Golgi apparatus, as well as ensuring a sufficient supply of UDP-GlcNAc (e.g., by expression of a UDP-GlcNAc transporter; see Examples below).
  • a GnT such as GnTI, GnTII, and GnTIII
  • nucleotide sugar precursors e.g., UDP-N-acetylglucosamine, UDP-N-acetylgalactosamine, CMP-N-acetylneuraminic acid, UDP-galactose, etc.
  • nucleotide sugar precursors e.g., UDP-N-acetylglucosamine, UDP-N-acetylgalactosamine, CMP-N-acetylneuraminic acid, UDP-galactose, etc.
  • sugar nucleoside specific transporters have to be expressed in the Golgi to ensure adequate levels of nucleoside sugar precursors (Sommers and Hirschberg (1981) J. Cell Biol. 91(2):A406-A406; Sommers and Hirschberg (1982) J. Biol. Chem. 257(18):811-817; Perez and Hirschberg (1987) Methods in Enzymology 138:709-715).
  • Nucleotide sugars may be provided to the appropriate compartments, e.g., by expressing in the host microorganism an exogenous gene encoding a sugar nucleotide transporter.
  • the choice of transporter enzyme is influenced by the nature of the exogenous glycosyltransferase being used.
  • a GlcNAc transferase may require a UDP-GlcNAc transporter
  • a fucosyltransferase may require a GDP-fucose transporter
  • a galactosyltransferase may require a UDP-galactose transporter
  • a sialyltransferase may require a CMP-sialic acid transporter.
  • the added transporter protein conveys a nucleotide sugar from the cytosol into the Golgi apparatus, where the nucleotide sugar may be reacted by the glycosyltransferase, e.g., to elongate an N-glycan.
  • the reaction liberates a nucleoside diphosphate or monophosphate, e.g., UDP, GDP, or CMP.
  • Nucleoside monophosphates can be directly exported from the Golgi in exchange for nucleoside triphosphate sugars by an antiport mechanism. Accumulation of a nucleoside diphosphate, however, inhibits the further activity of a glycosyltransferase.
  • the diphosphatase (specific for UDP or GDP as appropriate) hydrolyzes the diphosphonucleoside to yield a nucleoside monosphosphate and inorganic phosphate.
  • Suitable transporter enzymes which are typically of mammalian origin, are described below. Such enzymes may be engineered into a selected host cell using the methods of the invention.
  • ⁇ 2,3- or ⁇ 2,6-sialyltransferase caps galactose residues with sialic acid in the trans-Golgi and TGN of humans leading to a mature form of the glycoprotein ( FIG. 1B ).
  • this processing step into a metabolically engineered yeast or fungus will require (1) ⁇ 2,3- or ⁇ 2,6-sialyltransferase activity and (2) a sufficient supply of CMP-N-acetyl neuraminic acid, in the late Golgi of yeast.
  • the catalytic domain of a known sialyltransferase (e.g. from humans) has to be directed to the late Golgi in fungi (see above).
  • transporters have to be engineered to allow the transport of CMP-N-acetyl neuraminic acid into the late Golgi.
  • the cDNA of human UDP-N-acetylglucosamine transporter which was recognized through a homology search in the expressed sequence tags database (dbEST), has been cloned (Ishida (1999) J. Biochem. 126(1):68-77).
  • the mammalian Golgi membrane transporter for UDP-N-acetylglucosamine was cloned by phenotypic correction with cDNA from canine kidney cells (MDCK) of a recently characterized Kluyveromyces lactis mutant deficient in Golgi transport of the above nucleotide sugar (Guillen et al. (1998) Proc. Natl. Acad. Sci. USA 95(14):7888-7892).
  • a UDP-GlcNAc transporter in a host cell by means of a nucleic acid construct which may contain, for example: (1) a region by which the transformed construct is maintained in the cell (e.g., origin of replication or a region that mediates chromosomal integration), (2) a marker gene that allows for the selection of cells that have been transformed, including counterselectable and recyclable markers such as ura3 or T-urf13 (Soderholm et al.
  • the rat liver Golgi membrane GDP-fucose transporter has been identified and purified by Puglielli and Hirschberg (1999) J. Biol. Chem. 274(50):35596-35600.
  • the corresponding gene has not been identified, however, N-terminal sequencing can be used for the design of oligonucleotide probes specific for the corresponding gene. These oligonucleotides can be used as probes to clone the gene encoding for GDP-fucose transporter.
  • nucleoside diphosphate or monophosphate When sugars are transferred onto a glycoprotein, either a nucleoside diphosphate or monophosphate is released from the sugar nucleotide precursors. While monophosphates can be directly exported in exchange for nucleoside triphosphate sugars by an antiport mechanism, diphosphonucleosides (e.g. GDP) have to be cleaved by phosphatases (e.g. GDPase) to yield nucleoside monophosphates and inorganic phosphate prior to being exported. This reaction appears to be important for efficient glycosylation, as GDPase from S. cerevisiae has been found to be necessary for mannosylation. However, the enzyme only has 10% of the activity towards UDP (Berninsone et al.
  • UDP is known to be a potent inhibitor of glycosyltransferases and the removal of this glycosylation side product is important to prevent glycosyltransferase inhibition in the lumen of the Golgi (Khatara et al. (1974) Eur. J. Biochem. 44:537-560).
  • the present invention further provides a method for producing a human-like glycoprotein in a non-human host cell comprising the step of introducing into the cell one or more nucleic acid molecules which encode an enzyme or enzymes for production of the Man 5 GlcNAc 2 carbohydrate structure.
  • a nucleic acid molecule encoding one or more mannosidase activities involved in the production of Man 5 GlcNAc 2 from Man 8 GlcNAc 2 or Man 9 GlcNAc 2 is introduced into the host.
  • the invention additionally relates to methods for making altered glycoproteins in a host cell comprising the step of introducing into the host cell a nucleic acid molecule which encodes one or more glycosylation enzymes or activities.
  • Preferred enzyme activities are selected from the group consisting of UDP-GlcNAc transferase, UDP-galactosyltransferase, GDP-fucosyltransferase, CMP-sialyltransferase, UDP-GlcNAc transporter, UDP-galactose transporter, GDP-fucose transporter, CMP-sialic acid transporter, and nucleotide diphosphatases.
  • the host is selected or engineered to express two or more enzymatic activities in which the product of one activity increases substrate levels of another activity, e.g., a glycosyltransferase and a corresponding sugar transporter, e.g., GnTI and UDP-GlcNAc transporter activities.
  • the host is selected or engineered to expresses an activity to remove products which may inhibit subsequent glycosylation reactions, e.g. a UDP- or GDP-specific diphosphatase activity.
  • Preferred methods of the invention involve expressing one or more enzymatic activities from a nucleic acid molecule in a host cell and comprise the step of targeting at least one enzymatic activity to a desired subcellular location (e.g., an organelle) by forming a fusion protein comprising a catalytic domain of the enzyme and a cellular targeting signal peptide, e.g., a heterologous signal peptide which is not normally ligated to or associated with the catalytic domain.
  • a desired subcellular location e.g., an organelle
  • the fusion protein is encoded by at least one genetic construct (“fusion construct”) comprising a nucleic acid fragment encoding a cellular targeting signal peptide ligated in the same translational reading frame (“in-frame”) to a nucleic acid fragment encoding an enzyme (e.g., glycosylation enzyme), or catalytically active fragment thereof.
  • fusion construct comprising a nucleic acid fragment encoding a cellular targeting signal peptide ligated in the same translational reading frame (“in-frame”) to a nucleic acid fragment encoding an enzyme (e.g., glycosylation enzyme), or catalytically active fragment thereof.
  • the targeting signal peptide component of the fusion construct or protein is preferably derived from a member of the group consisting of: membrane-bound proteins of the ER or Golgi, retrieval signals, Type II membrane proteins, Type I membrane proteins, membrane spanning nucleotide sugar transporters, mannosidases, sialyltransferases, glucosidases, mannosyltransferases and phosphomannosyltransferases.
  • the catalytic domain component of the fusion construct or protein is preferably derived from a glycosidase, mannosidase or a glycosyltransferase activity derived from a member of the group consisting of GnTI, GnTII, GnTIII, GnTIV, GnTV, GnTVI, GalT, Fucosyltransferase and Sialyltransferase.
  • the catalytic domain preferably has a pH optimum within 1.4 pH units of the average pH optimum of other representative enzymes in the organelle in which the enzyme is localized, or has optimal activity at a pH between 5.1 and 8.0.
  • the catalytic domain encodes a mannosidase selected from the group consisting of C.
  • elegans mannosidase IA C. elegans mannosidase IB, D. melanogaster mannosidase IA, H. sapiens mannosidase IB, P. citrinum mannosidase I, mouse mannosidase IA, mouse mannosidase IB, A. nidulans mannosidase IA, A. nidulans mannosidase IB, A. nidulans mannosidase IC, mouse mannosidase II, C. elegans mannosidase II, H. sapiens mannosidase II, mannosidase Iix, and mannosidase III.
  • a human-like glycoprotein is made efficiently in a non-human eukaryotic host cell by introducing into a subcellular compartment of the cell a glycosylation enzyme selected to have a pH optimum similar to the pH optima of other enzymes in the targeted subcellular compartment.
  • a glycosylation enzyme selected to have a pH optimum similar to the pH optima of other enzymes in the targeted subcellular compartment.
  • most enzymes that are active in the ER and Golgi apparatus of S. cerevisiae have pH optima that are between about 6.5 and 7.5 (see Table 3). Because the glycosylation of proteins is a highly evolved and efficient process, the internal pH of the ER and the Golgi is likely also in the range of about 6-8.
  • a preferred embodiment of this invention targets a selected glycosylation enzyme (or catalytic domain thereof), e.g., an ⁇ -mannosidase, to a subcellular location in the host cell (e.g., an organelle) where the pH optimum of the enzyme or domain is within 1.4 pH units of the average pH optimum of other representative marker enzymes localized in the same organelle(s).
  • the pH optimum of the enzyme to be targeted to a specific organelle should be matched with the pH optimum of other enzymes found in the same organelle to maximize the activity per unit enzyme obtained.
  • Table 3 summarizes the activity of mannosidases from various sources and their respective pH optima.
  • Table 4 summarizes their typical subcellular locations.
  • a particular enzyme or catalytic domain is targeted to a subcellular location in the host cell by means of a chimeric fusion construct encoding a protein comprising a cellular targeting signal peptide not normally associated with the enzymatic domain.
  • an enzyme or domain is targeted to the ER, the early, medial or late Golgi, or the trans Golgi apparatus of the host cell.
  • the targeted glycosylation enzyme is a mannosidase, glycosyltransferase or a glycosidase.
  • mannosidase activity is targeted to the ER or cis Golgi, where the early reactions of glycosylation occur. While this method is useful for producing a human-like glycoprotein in a non-human host cell, it will be appreciated that the method is also useful more generally for modifying carbohydrate profiles of a glycoprotein in any eukaryotic host cell, including human host cells.
  • Targeting sequences which mediate retention of proteins in certain organelles of the host cell secretory pathway are well-known and described in the scientific literature and public databases, as discussed in more detail below with respect to libraries for selection of targeting sequences and targeted enzymes.
  • Such subcellular targeting sequences may be used alone or in combination to target a selected glycosylation enzyme (or catalytic domain thereof) to a particular subcellular location in a host cell, i.e., especially to one where the enzyme will have enhanced or optimal activity based on pH optima or the presence of other stimulatory factors.
  • Man 5 GlcNAc 2 When one attempts to trim high mannose structures to yield Man 5 GlcNAc 2 in the ER or the Golgi apparatus of a host cell such as S. cerevisiae , for example, one may choose any enzyme or combination of enzymes that (1) has a sufficiently close pH optimum (i.e., between pH 5.2 and pH 7.8), and (2) is known to generate, alone or in concert, the specific isomeric Man 5 GlcNAc 2 structure required to accept subsequent addition of GlcNAc by GnTI. Any enzyme or combination of enzymes that is shown to generate a structure that can be converted to GlcNAcMan 5 GlcNAc 2 by GnTI in vitro would constitute an appropriate choice. This knowledge may be obtained from the scientific literature or experimentally.
  • Mannosidase can convert Man 8 GlcNAc 2 -2AB (2-aminobenzamide) to Man 5 GlcNAc 2 -AB and then verify that the obtained Man 5 GlcNAc 2 -2AB structure can serve a substrate for GnTI and UDP-GlcNAc to give GlcNAcMan 5 GlcNAc 2 in vitro.
  • Mannosidase IA from a human or murine source, for example, would be an appropriate choice (see, e.g., Example 4). Examples described herein utilize 2-aminobenzamide labeled N-linked oligomannose followed by HPLC analysis to make this determination.
  • a glycosylation enzyme such as an ⁇ -1,2-mannosidase enzyme used according to the invention has an optimal activity at a pH of between 5.1 and 8.0.
  • the enzyme has an optimal activity at a pH of between 5.5 and 7.5.
  • the C. elegans mannosidase enzyme works well in the methods of the invention and has an apparent pH optimum of about 5.5).
  • Preferred mannosidases include those listed in Table 3 having appropriate pH optima, e.g.
  • Aspergillus nidulans Homo sapiens IA (Golgi), Homo sapiens IB (Golgi), Lepidopteran insect cells (IPLB-SF21 AE), Homo sapiens, mouse IB (Golgi), Xanthomonas manihotis, Drosophila melanogaster and C. elegans.
  • Example 7 An experiment which illustrates the pH optimum for an ⁇ -1,2-mannosidase enzyme is described in Example 7.
  • a chimeric fusion protein BB27-2 Saccharomyces MNN10 (s)/ C. elegans mannosidase IB ⁇ 31), which leaks into the medium was subjected to various pH ranges to determine the optimal activity of the enzyme.
  • the results of the experiment show that the ⁇ -1,2-mannosidase has an optimal pH of about 5.5 for its function ( FIG. 11 ).
  • a single cloned mannosidase gene is expressed in the hostlorganism.
  • the encoded mannosidases preferably all have pH optima within the preferred range of about 5.1 to about 8.0, or especially between about 5.5 and about 7.5.
  • Preferred mannosidase activities include ⁇ -1,2-mannosidases derived from mouse, human, Lepidoptera, Aspergillus nidulans, or Bacillus sp., C. elegans, D. melanogaster, P. citrinum, X laevis or A. nidulans.
  • Certain methods of the invention are preferably (but need not necessarily be) carried out using one or more nucleic acid libraries.
  • An exemplary feature of a combinatorial nucleic acid library of the invention is that it comprises sequences encoding cellular targeting signal peptides and sequences encoding proteins to be targeted (e.g., enzymes or catalytic domains thereof, including but not limited to those which mediate glycosylation).
  • a combinatorial nucleic acid library comprises: (a) at least two nucleic acid sequences encoding different cellular targeting signal peptides; and (b) at least one nucleic acid sequence encoding a polypeptide to be targeted.
  • a combinatorial nucleic acid library comprises: (a) at least one nucleic acid sequence encoding a cellular targeting signal peptide; and (b) at least two nucleic acid sequences encoding a polypeptide to be targeted into a host cell.
  • a nucleic acid sequence derived from (a) and a nucleic acid sequence derived from (b) are ligated to produce one or more fusion constructs encoding a cellular targeting signal peptide functionally linked to a polypeptide domain of interest.
  • a functional linkage is when the cellular targeting signal peptide is ligated to the polypeptide domain of interest in the same translational reading frame (“in-frame”).
  • a combinatorial DNA library expresses one or more fusion proteins comprising cellular targeting signal peptides ligated in-frame to catalytic enzyme domains.
  • the encoded fusion protein preferably comprises a catalytic domain of an enzyme involved in mammalian- or human-like modification of N-glycans.
  • the catalytic domain is derived from an enzyme selected from the group consisting of mannosidases, glycosyltransferases and other glycosidases which is ligated in-frame to one or more targeting signal peptides.
  • the enzyme domain may be exogenous and/or endogenous to the host cell.
  • a particularly preferred signal peptide is one normally associated with a protein that undergoes ER to Golgi transport.
  • the combinatorial DNA library of the present invention may be used for producing and localizing in vivo enzymes involved in mammalian- or human-like N-glycan modification.
  • the fusion constructs of the combinatorial DNA library are engineered so that the encoded enzymes are localized in the ER, Golgi or the trans-Golgi network of the host cell where they are involved in producing particular N-glycans on a glycoprotein of interest.
  • Localization of N-glycan modifying enzymes of the present invention is achieved through an anchoring mechanism or through protein-protein interaction where the localization peptide constructed from the combinatorial DNA library localizes to a desired organelle of the secretory pathway such as the ER, Golgi or the trans Golgi network.
  • Man 5 GlcNAc 2 An example of a useful N-glycan, which is produced efficiently and in sufficient quantities for further modification by human-like (complex) glycosylation reactions is Man 5 GlcNAc 2 .
  • a sufficient amount of Man 5 GlcNAc 2 is needed on a glycoprotein of interest for further human-like processing in vivo (e.g., more than 30 mole %).
  • the Man 5 GlcNAc 2 intermediate may be used as a substrate for further N-glycan modification to produce GlcNAcMan 5 GlcNAc 2 ( FIG. 1B ; see above).
  • the combinatorial DNA library of the present invention may be used to produce enzymes that subsequently produce GlcNAcMan 5 GlcNAc 2 , or other desired complex N-glycans, in a useful quantity.
  • a further aspect of the fusion constructs produced using the combinatorial DNA library of the present invention is that they enable sufficient and often near complete intracellular N-glycan trimming activity in the engineered host cell.
  • Preferred fusion constructs produced by the combinatorial DNA library of the invention encode a glycosylation enzyme, e.g., a mannosidase, which is effectively localized to an intracellular host cell compartment and thereby exhibits very little and preferably no extracellular activity.
  • the preferred fusion constructs of the present invention that encode a mannosidase enzyme are shown to localize where the N-glycans are modified, namely, the ER and the Golgi.
  • the fusion enzymes of the present invention are targeted to such particular organelles in the secretory pathway where they localize and act upon N-glycans such as Man 8 GlcNAc 2 to produce Man 5 GlcNAc 2 on a glycoprotein of interest.
  • GnTIII fusion constructs generated from a combinatorial DNA library to produce bisected glycans were assayed to determine any extracellular activity:
  • An example of a GnTIII fusion constructs exhibiting in vivo alteration of host cell glycosylation is designated pVA53.
  • the supernatant was tested to detect any ex vivo GnTIII activity.
  • FIG. 33 shows no apparent change in the standard substrate GlcNAcMan 5 GlcNAc 2 under conditions that would reveal extracellular GnTIII activity in the medium (Example 22).
  • FIG. 34 shows no detectable extracellular GnTIII activity in the medium in P. pastoris YSH-57 reacting with the substrate GlcNAc 2 Man 3 GlcNAc 2 (Example 23).
  • Enzymes produced by the combinatorial DNA library of the present invention can modify N-glycans on a glycoprotein of interest as shown for K3 or IFN- ⁇ proteins expressed in P. pastoris , as shown in FIGS. 5, 6 , and 25 - 34 (see also Examples 2, 4, and 18-23).
  • glycoproteins without limitation, including erythropoietin, cytokines such as interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , and granulocyte-CSF, coagulation factors such as factor VIII, factor IX, and human protein C, soluble IgE receptor ⁇ -chain, IgG, IgG fragments, IgM, urokinase, chymase, urea trypsin inhibitor, IGF-binding protein, epidermal growth factor, growth hormone-releasing factor, annexin V fusion protein, angiostatin, vascular endothelial growth factor-2, myeloid progenitor inhibitory factor-1, osteoprotegerin, ⁇ -1 antitrypsin, DNase II, and ⁇ -feto proteins may be glycosylated in this way.
  • cytokines such as interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , and granulocyte-C
  • a combinatorial DNA library of fusion constructs features one or more cellular targeting signal peptides (“targeting peptides”) generally derived from N-terminal domains of native proteins (e.g., by making C-terminal deletions). Some targeting peptides, however, are derived from the C-terminus of native proteins (e.g. SEC12).
  • Membrane-bound proteins of the ER or the Golgi are preferably used as a source for targeting peptide sequences. These proteins have sequences encoding a cytosolic tail (ct), a transmembrane domain (tmd) and a stem region (sr) which are varied in length. These regions are recognizable by protein sequence alignments and comparisons with known homologs and/or other localized proteins (e.g., comparing hydrophobicity plots).
  • the targeting peptides are indicated herein as short (s), medium (m) and long (1) relative to the parts of a type II membrane.
  • the targeting peptide sequence indicated as short (s) corresponds to the transmembrane domain (tmd) of the membrane-bound protein.
  • the targeting peptide sequence indicated as long (1) corresponds to the length of the transmembrane domain (tmd) and the stem region (sr).
  • the targeting peptide sequence indicated as medium (m) corresponds to the transmembrane domain (tmd) and approximately half the length of the stem region (sr).
  • the catalytic domain regions are indicated herein by the number of nucleotide deletion with respect to its wild-type glycosylation enzyme.
  • a combinatorial nucleic acid library of the invention may be assembled directly from existing or wild-type genes.
  • the DNA library is assembled from the. fusion of two or more sub-libraries. By the in-frame ligation of the sub-libraries, it is possible to create a large number of novel genetic constructs encoding useful targeted protein domains such as those which have glycosylation activities.
  • One useful sub-library includes DNA sequences encoding enzymes such as glycosidases (e.g., mannosidases), glycosyltransferases (e.g., fucosyl-transferases, galactosyltransferases, glucosyltransferases), GlcNAc transferases and sialyltransferases.
  • Catalytic domains may be selected from the host to be engineered, as well as from other related or unrelated organisms. Mammalian, plant, insect, reptile, algal or fungal enzymes are all useful and should be chosen to represent a broad spectrum of biochemical properties with respect to temperature and pH optima.
  • genes are truncated to give fragments some of which encode the catalytic domains of the enzymes. By removing endogenous targeting sequences, the enzymes may then be redirected and expressed in other cellular loci.
  • catalytic domains may be guided by the knowledge of the particular environment in which the catalytic domain is subsequently to be active. For example, if a particular glycosylation enzyme is to be active in the late Golgi, and all known enzymes of the host organism in the late Golgi have a certain pH optimum, or the late Golgi is known to have a particular pH, then a catalytic domain is chosen which exhibits adequate, and preferably maximum, activity at that pH, as discussed above.
  • Another useful sub-library includes nucleic acid sequences encoding targeting signal peptides that result in localization of a protein to a particular location within the ER, Golgi, or trans Golgi network. These targeting peptides may be selected from the host organism to be engineered as well as from other related or unrelated organisms.
  • N-terminal sequences encoding a cytosolic tail (ct), a transmembrane domain (tmd) and part or all of a stem region (sr), which together or individually anchor proteins to the inner (lumenal) membrane of the Golgi; (2) retrieval signals which are generally found at the C-terminus such as the HDEL (SEQ ID NO:41) or KDEL (SEQ ID NO:42) tetrapeptide; and (3) membrane spanning regions from various proteins, e.g., nucleotide sugar transporters, which are known to localize in the Golgi.
  • the library is designed such that the ct, the tmd and various parts of the stem region are represented.
  • a preferred embodiment of the sub-library of targeting peptide sequences includes ct, tmd, and/or sr sequences from membrane-bound proteins of the ER or Golgi.
  • Still other useful sources of targeting peptide sequences include retrieval signal peptides, e.g. the tetrapeptides HDEL or KDEL, which are typically found at the C-terminus of proteins that are transported retrograde into the ER or Golgi.
  • Still other sources of targeting peptide sequences include (a) type II membrane proteins, (b) the enzymes listed in Table 3, (c) membrane spanning nucleotide sugar transporters that are localized in the Golgi, and (d) sequences referenced in Table 5.
  • TABLE 5 Sources of useful compartmental targeting sequences Gene or Location of Sequence Organism Function Gene Product MNSI A. nidulans ⁇ -1,2-mannosidase ER MNSI A.
  • niger ⁇ -1,2-mannosidase ER MNSI S. cerevisiae ⁇ -1,2-mannosidase ER GLSI S. cerevisiae glucosidase ER GLSI A. niger glucosidase ER GLSI A. nidulans glucosidase ER HDEL Universal in retrieval signal ER at C- fungi terminus SEC12 S. cerevisiae COPII vesicle protein ER/Golgi SEC12 A. niger COPII vesicle protein ER/Golgi OCH1 S.
  • targeting peptide sequences are selected which are appropriate for the particular enzymatic activity or activities to function optimally within the sequence of desired glycosylation reactions.
  • a process which occurs in the late Golgi in humans it is desirable to utilize a sub-library of targeting peptide sequences derived from late Golgi proteins.
  • the trimming of Man 8 GlcNAc 2 by an ⁇ -1,2-mannosidase to give Man 5 GlcNAc 2 is an early step in complex N-glycan formation in humans ( FIG. 1B ). It is therefore desirable to have this reaction occur in the ER or early Golgi of an engineered host microorganism.
  • a sub-library encoding ER and early Golgi retention signals is used.
  • a series of fusion protein constructs (i.e., a combinatorial DNA library) is then constructed by functionally linking one or a series of targeting peptide sequences to one or a series of sequences encoding catalytic domains.
  • this is accomplished by the in-frame ligation of a sub-library comprising DNA encoding targeting peptide sequences (above) with a sub-library comprising DNA encoding glycosylation enzymes or catalytically active fragments thereof (see below).
  • the resulting library comprises synthetic genes encoding targeting peptide sequence-containing fusion proteins.
  • a targeting peptide sequence at the N-terminus of a fusion protein, or in other cases at the C-terminus.
  • targeting peptide sequences may be inserted within the open reading frame of an enzyme, provided the protein structure of individual folded domains is not disrupted.
  • Each type of fusion protein is constructed (in a step-wise directed or semi-random fashion) and optimal constructs may be selected upon transformation of host cells and characterization of glycosylation patterns in transformed cells using methods of the invention.
  • the construction of a preferred combinatorial DNA library is illustrated schematically in FIG. 2 and described in Example 4.
  • the fusion construct may be operably linked to a multitude of vectors, such as expression vectors well-known in the art.
  • a wide variety of such fusion constructs were assembled using representative activities as shown in Table 6.
  • Combinations of targeting peptide/catalytic domains may be assembled for use in targeting mannosidase, glycosyltransferase and glycosidase activities in the ER, Golgi, and the trans Golgi network according to the invention.
  • the same catalytic domain may have no effect to a very profound effect on N-glycosylation patterns, depending on the type of targeting peptide used (see, e.g., Table 7, Example 4).
  • a representative example of a mannosidase fusion construct derived from a combinatorial DNA library of the invention is pFB8, which a truncated Saccharomyces SEC12(m) targeting peptide (988-1296 nucleotides of SEC12 from SwissProt P11655) ligated in-frame to a 187 N-terminal amino acid deletion of a mouse ⁇ -mannosidase IA (Genbank AN 6678787).
  • the encoded fusion protein localizes in the ER by means of the SEC12 targeting peptide sequence while retaining its mannosidase catalytic domain activity and is capable of producing in vivo N-glycans having a Man 5 GlcNAc 2 structure (Example 4; FIGS. 6F and 7B ).
  • the fusion construct pGC5, Saccharomyces MNS1 (m)/mouse mannosidase IB ⁇ 99 is another example of a fusion construct having intracellular mannosidase trimming activity (Example 4; FIGS. 5D and 8B ).
  • Fusion construct pBC18-5 Saccharomyces VAN1 (s)/ C. elegans mannosidase IB ⁇ 80
  • mannosidase trimming activity may be specific to a particular protein of interest.
  • a protein of interest may be introduced into a host cell transfected with a combinatorial DNA library to identify one or more fusion constructs which express a mannosidase activity optimal for the protein of interest.
  • One skilled in the art will be able to produce and select optimal fusion construct(s) using the combinatorial DNA library approach described herein.
  • glycosyltransferase combinatorial DNA library was made using the methods of the invention.
  • a combinatorial DNA library of sequences derived from glycosyltransferase I (GnTI) activities were assembled with targeting peptides and screened for efficient production in a lower eukaryotic host cell of a GlcNAcMan 5 GlcNAc 2 N-glycan structure on a marker glycoprotein.
  • GnTI glycosyltransferase I
  • a fusion construct shown to produce GlcNAcMan 5 GlcNAc 2 (pPB1O4), Saccharomyces MNN9(s)/human GnTI A38 was identified (Example 8).
  • a wide variety of such GnTI fusion constructs were assembled (Example 8, Table 10).
  • targeting peptide/GnTI catalytic domains can readily be assembled by making a combinatorial DNA library. It is also apparent to one skilled in the art that other such fusion constructs exhibiting glycosyltransferase activity may be made as demonstrated in Example 8. It will be a matter of routine experimentation for one skilled in the art to use the combinatorial DNA library method described herein to optimize GlcNAcMan 5 GlcNAc 2 production using a selected fusion construct in a particular expression vector and host cell line.
  • Example 8 illustrates a preferred embodiment of a combinatorial DNA library comprising targeting peptides and GnTI catalytic domain fusion constructs involved in producing glycoproteins with predominantly GlcNAcMan 5 GlcNAc 2 structure.
  • a P. pastoris strain with an OCH1 deletion that expresses a reporter protein (K3) was transformed with multiple fusion constructs isolated from combinatorial libraries of the invention to convert high mannose N-glycans to human-like N-glycans (Example 8).
  • the mannosidase fusion construct pFB8 Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187
  • pPB103 comprising a K.
  • lactis MNN2-2 gene (Genbank AN AF 106080) encoding an UDP-GlcNAc transporter was constructed to increase further production of GlcNAcMan 5 GlcNAc 2 .
  • the addition of the UDP-GlcNAc transporter increased production of GlcNAcMan 5 GlcNAc 2 significantly in the P. pastoris strain as illustrated in FIG. 10B .
  • pPB104 comprising Saccharomyces MNN9 (s)/human GnTI ⁇ 38 was introduced into the strain. This P. pastoris strain is referred to as “PBP-3.” (See FIG. 36 .)
  • host cells such as the above-described yeast strains can be sequentially transformed and/or co-transformed with one or more expression vectors. It is also understood that the order of transformation is not particularly relevant in producing the glycoprotein of interest. The skilled artisan recognizes the routine modifications of the procedures disclosed herein may provide improved results in the production of the glycoprotein of interest.
  • the combinatorial DNA library provides a tool for constructing enzyme fusions that are involved in modifying N-glycans on a glycoprotein of interest, which is especially useful in producing human-like glycoproteins.
  • Any enzyme fusion may be selected using libraries and methods of the invention.
  • Desired transformants expressing appropriately targeted, active ⁇ -1,2-mannosidase produce K3 with N-glycans of the structure Man 5 GlcNAc 2 as shown in FIGS. 5D and 5E . This confers a reduced molecular mass to the cleaved glycan compared to the K3 of the parent OCH1 deletion strain, as was detected by MALDI-TOF mass spectrometry in FIG. 5C .
  • IFN- ⁇ comprising predominantly Man 5 GlcNAc 2 .
  • the Man 5 GlcNAc 2 was removed by PNGase digestion (Papac et al. (1998) Glycobiology 8:445-454) and subjected to MALDI-TOF as shown in FIGS. 6A-6F .
  • a single prominent peak at 1254 (m/z) confirms Man 5 GlcNA 2 production on IFN- ⁇ in FIGS.
  • 6E (pGC5) ( Saccharomyces MNS1 (m)/mouse mannosidase IB ⁇ 99) and 6 F (pFB8) ( Saccharomyces SEC 12 (m)/mouse mannosidase IA ⁇ 187).
  • pFB8 Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187
  • pPB104 Saccharomyces MNN9 (s)/human GnTI ⁇ 38
  • pPB103 K. lactis MNN2-2 gene
  • the MNN1 gene was cloned from K. lactis as described in Example 9.
  • the nucleic acid and deduced amino acid sequences of the K. lactis MNN1 gene are shown in SEQ ID NOs:43 and 44, respectively.
  • a construct was made to delete the MNN1 gene from the genome of K. lactis (Example 9).
  • Host cells depleted in och1 and mnn1 activities produce N-glycans having a Man 9 GlcNAc 2 carbohydrate structure (see, e.g., FIG. 12B ).
  • Such host cells may be engineered further using, e.g., methods and libraries of the invention, to produce mammalian- or human-like glycoproteins.
  • the invention provides an isolated nucleic acid molecule having a nucleic acid sequence comprising or consisting of at least forty-five, preferably at least 50, more preferably at least 60 and most preferably 75 or more nucleotide residues of the K. lactis MNN1 gene (SEQ ID NO: 43), and homologs, variants and derivatives thereof.
  • the invention also provides nucleic acid molecules that hybridize under stringent conditions to the above-described nucleic acid molecules.
  • isolated polypeptides including muteins, allelic variants, fragments, derivatives, and analogs
  • vectors including expression vectors, which comprise a nucleic acid molecule of the invention, as described further herein.
  • host cells transformed with the nucleic acid molecules or vectors of the invention are provided.
  • Another aspect of the present invention thus relates to a non-human eukaryotic host strain expressing glycoproteins comprising modified N-glycans that resemble those made by human-cells. Performing the methods of the invention in species other than yeast and fungal cells is thus contemplated and encompassed by this invention. It is contemplated that a combinatorial nucleic acid library of the present invention may be used to select constructs that modify the glycosylation pathway in any eukaryotic host cell system.
  • the combinatorial libraries of the invention may also be used in plants, algae and insects, and in other eukaryotic host cells, including mammalian and human cells, to localize proteins, including glycosylation enzymes or catalytic domains thereof, in a desired location along a host cell secretory pathway.
  • glycosylation enzymes or catalytic domains and the like are targeted to a subcellular location along the host cell secretory pathway where they are capable of functioning, and preferably, where they are designed or selected to function most efficiently.
  • Plant and insect cells may also be engineered to alter the glycosylation of expressed proteins using the combinatorial library and methods of the invention.
  • glycosylation in mammalian cells, including human cells may also be modified using the combinatorial library and methods of the invention. It may be possible, for example, to optimize a particular enzymatic activity or to otherwise modify the relative proportions of various N-glycans made in a mammalian host cell using the combinatorial library and methods of the invention.
  • Examples of modifications to glycosylation which can be affected using a method according to this embodiment of the invention are: (1) engineering a eukaryotic host cell to trim mannose residues from Man 8 GlcNAc 2 to yield a Man 5 GlcNAc 2 N-glycan; (2) engineering eukaryotic host cell to add an N-acetylglucosamine (GlcNAc) residue to Man 5 GlcNAc 2 by action of GlcNAc transferase I; (3) engineering.a eukaryotic host cell to functionally express an enzyme such as an N-acetylglucosaminyl Transferase (GnTI, GnTII, GnTIII, GnTIV, GnTV, GnTVI), mannosidase II, fucosyltransferase (FT), galactosyl tranferase (GalT) or a sialyltransferase (ST).
  • a target host such as a lower eukaryotic microorganism.
  • the host organism is transformed two or more times with DNA libraries including sequences encoding glycosylation activities. Selection of desired phenotypes may be performed after each round of transformation or alternatively after several transformations have occurred. Complex glycosylation pathways can be rapidly engineered in this manner.
  • such targeting peptide/catalytic domain libraries are designed to incorporate existing information on the sequential nature of glycosylation reactions in higher eukaryotes. Reactions known to occur early in the course of glycoprotein processing require the targeting of enzymes that catalyze such reactions to an early part of the Golgi or the ER. For example, the trimming of Man 8 GlcNAc 2 to Man 5 GlcNAc 2 by mannosidases is an early step in complex N-glycan formation ( FIGS. 1B and 35A ). Because protein processing is initiated in the ER and then proceeds through the early, medial and late Golgi, it is desirable to have this reaction occur in the ER or early Golgi. When designing a library for mannosidase I localization, for example, one thus attempts to match ER and early Golgi targeting signals with the catalytic domain of mannosidase I.
  • the method of this embodiment is most effective when a nucleic acid, e.g., a DNA library transformed into the host contains a large diversity of sequences, thereby increasing the probability that at least one transformant will exhibit the desired phenotype.
  • Single amino acid mutations may drastically alter the activity of glycoprotein processing enzymes (Romero et al. (2000) J. Biol. Chem. 275(15):11071-4).
  • a DNA library or a constituent sub-library may be subjected to one or more techniques to generate additional sequence diversity. For example, one or more rounds of gene shuffling, error prone PCR, in vitro mutagenesis or other methods for generating sequence diversity, may be performed to obtain a larger diversity of sequences within the pool of fusion constructs.
  • each library construct with expression control sequences, such as promoters, transcription terminators, enhancers, ribosome binding sites, and other functional sequences as may be necessary to ensure effective transcription and translation of the fusion proteins upon transformation of fusion constructs into the host organism.
  • expression control sequences such as promoters, transcription terminators, enhancers, ribosome binding sites, and other functional sequences as may be necessary to ensure effective transcription and translation of the fusion proteins upon transformation of fusion constructs into the host organism.
  • Suitable vector components e.g., selectable markers, expression control sequences (e.g., promoter, enhancers, terminators and the like) and, optionally, sequences required for autonomous replication in a host cell, are selected as a function of which particular host cell is chosen. Selection criteria for suitable vector components for use in a particular mammalian or a lower eukaryotic host cell are routine.
  • Preferred lower eukaryotic host cells of the invention include Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium lucknowense, Fusarium sp.
  • suitable promoters include, for example, the AOX1, AOX2, GAPDH and P40 promoters.
  • each construct with at least one selectable marker, such as a gene to impart drug resistance or to complement a host metabolic lesion.
  • selectable marker such as a gene to impart drug resistance or to complement a host metabolic lesion.
  • the presence of the marker is useful in the subsequent selection of transformants; for example, in yeast the URA3, HIS4, SUC2, G418, BLA, or SH BLE genes may be used.
  • a multitude of selectable markers are known and available for use in yeast, fungi, plant, insect, mammalian and other eukaryotic host cells.
  • the nucleic acid library is then transformed into the host organism.
  • yeast any convenient method of DNA transfer may be used, such as electroporation, the lithium chloride method, or the spheroplast method.
  • filamentous fungi and plant cells conventional methods include particle bombardment, electroporation and agrobacterium mediated transformation.
  • To produce a stable strain suitable for high-density culture e.g., fermentation in yeast, it is desirable to integrate the DNA library constructs into the host chromosome.
  • integration occurs via homologous recombination, using techniques well-known in the art.
  • DNA library elements are provided with flanking sequences homologous to sequences of the host organism. In this manner, integration occurs at a defined site in the host genome, without disruption of desirable or essential genes.
  • library DNA is integrated into the site of an undesired gene in a host chromosome, effecting the disruption or deletion of the gene.
  • integration into the sites of the OCH1, MNN1, or MNN4 genes allows the expression of the desired library DNA while preventing the expression of enzymes involved in yeast hypermannosylation of glycoproteins.
  • library DNA may be introduced into the host via a nucleic acid molecule, plasmid, vector (e.g., viral or retroviral vector), chromosome, and may be introduced as an autonomous nucleic acid molecule or by homologous or random integration into the host genome.
  • Recyclable marker genes such as ura3, which can be selected for or against, are especially suitable.
  • transformants displaying a desired glycosylation phenotype are selected. Selection may be performed in a single step or by a series of phenotypic enrichment and/or depletion steps using any of a variety of assays or detection methods. Phenotypic characterization may be carried out manually or using automated high-throughput screening equipment. Commonly, a host microorganism displays protein N-glycans on the cell surface, where various glycoproteins are localized.
  • Such a screen may be based on a visual method, like a staining procedure, the ability to bind specific terminal GlcNAc binding antibodies or lectins conjugated to a marker (such lectins are available from E.Y.
  • FACS Fluorescence Assisted Cell Sorting
  • intact cells may be screened for a desired glycosylation phenotype by exposing the cells to a lectin or antibody that binds specifically to the desired N-glycan.
  • a lectin or antibody that binds specifically to the desired N-glycan.
  • a wide variety of oligosaccharide-specific lectins are available commercially (e.g., from EY Laboratories, San Mateo, Calif.).
  • antibodies to specific human or animal N-glycans are available commercially or may be produced using standard techniques.
  • An appropriate lectin or antibody may be conjugated to a reporter molecule, such as a chromophore, fluorophore, radioisotope, or an enzyme having a chromogenic substrate (Guillen et al., 1998. Proc. Natl. Acad. Sci. USA 95(14): 7888-7892).
  • Screening may then be performed using analytical methods such as spectrophotometry, fluorimetry, fluorescence activated cell sorting, or scintillation counting. In other cases, it may be necessary to analyze isolated glycoproteins or N-glycans from transformed cells. Protein isolation may be carried out by techniques known in the art. In a preferred embodiment, a reporter protein is secreted into the medium and purified by affinity chromatography (e.g. Ni-affinity or glutathione-S-transferase affinity chromatography).
  • affinity chromatography e.g. Ni-affinity or glutathione-S-transferase affinity chromatography
  • an enzyme such as endo- ⁇ -N-acetylglucosaminidase (Genzyme Co., Boston, Mass.; New England Biolabs, Beverly, Mass.) may be used to cleave the N-glycans from glycoproteins. Isolated proteins or N-glycans may then be analyzed by liquid chromatography (e.g., HPLC), mass spectroscopy, or other suitable means.
  • liquid chromatography e.g., HPLC
  • mass spectroscopy e.g., mass spectroscopy, or other suitable means.
  • U.S. Pat. No. 5,595,900 teaches several methods by which cells with desired extracellular carbohydrate structures may be identified.
  • MALDI-TOF mass spectrometry is used to analyze the cleaved N-glycans.
  • the desired transformants Prior to selection of a desired transformant, it may be desirable to deplete the transformed population of cells having undesired phenotypes. For example, when the method is used to engineer a functional mannosidase activity into cells, the desired transformants will have lower levels of mannose in cellular glycoprotein. Exposing the transformed population to a lethal radioisotope of mannose in the medium depletes the population of transformants having the undesired phenotype, i.e., high levels of incorporated mannose (Huffaker and Robbins (1983) Proc Natl Acad Sci USA. 80(24):7466-70).
  • a cytotoxic lectin or antibody, directed against an undesirable N-glycan may be used to deplete a transformed population of undesired phenotypes (e.g., Stanley and Siminovitch (1977) Somatic Cell Genet 3(4):391-405).
  • U.S. Pat. No. 5,595,900 teaches several methods by which cells with a desired extracellular carbohydrate structures may be identified. Repeatedly carrying out this strategy allows for the sequential engineering of more and more complex glycans in lower eukaryotes.
  • This screen may be carried out by growing cells harboring the transformed library under selective pressure on an agar plate and transferring individual colonies into a 96-well microtiter plate. After growing the cells, the cells are centrifuged, the cells resuspended in buffer, and after addition of UDP-GlcNAc and GnTII, the release of UDP is determined either by HPLC or an enzyme linked assay for UDP.
  • radioactively labeled UDP-GlcNAc and GnTII wash the cells and then look for the release of radioactive GlcNAc by N-actylglucosaminidase. All this may be carried manually or automated through the use of high throughput screening equipment. Transformants that release more UDP, in the first assay, or more radioactively labeled GlcNAc in the second assay, are expected to have a higher degree of GlcNAcMan 3 GlcNAc 2 on their surface and thus constitute the desired phenotype. Similar assays may be adapted to look at the N-glycans on secreted proteins as well.
  • any other suitable screen such as a lectin binding assay that is able to reveal altered glycosylation patterns on the surface of transformed cells.
  • the reduced binding of lectins specific to terminal mannoses may be a suitable selection tool.
  • Galantus nivalis lectin binds specifically to terminal ⁇ -1,3 mannose, which is expected to be reduced if sufficient mannosidase II activity is present in the Golgi.
  • This separation step would be carried out with a lectin column that specifically binds cells with a high terminal mannose content (e.g., Galantus nivalis lectin bound to agarose, Sigma, St.Louis, Mo.) over those that have a low terminal mannose content.
  • a lectin column that specifically binds cells with a high terminal mannose content (e.g., Galantus nivalis lectin bound to agarose, Sigma, St.Louis, Mo.) over those that have a low terminal mannose content.
  • the integration of multiple genes into the host (e.g., fungal) chromosome preferably involves careful planning.
  • the engineered strain may likely have to be transformed with a range of different genes, and these genes will have to be transformed in a stable fashion to ensure that the desired activity is maintained throughout the fermentation process.
  • any combination of various desired enzyme activities may be engineered into the fungal protein expression host, e.g., sialyltransferases, mannosidases, fucosyltransferases, galactosyltransferases, glucosyltransferases, GlcNAc transferases, ER and Golgi specific transporters (e.g. syn and antiport transporters for UDP-galactose and other precursors), other enzymes involved in the processing of oligosaccharides, and enzymes involved in the synthesis of activated oligosaccharide precursors such as UDP-galactose, CMP-N-acetylneuraminic acid.
  • sialyltransferases mannosidases
  • fucosyltransferases fucosyltransferases
  • galactosyltransferases galactosyltransferases
  • glucosyltransferases glucosyltransferases
  • Examples of preferred methods for modifying glycosylation in a lower eukaryotic host cell, such as Pichia pastoris are shown in Table 6. TABLE 6 Some preferred embodiments for modifying glycosylation in a lower eukaroytic microorganism Suitable Suitable Suitable Sources of Suitable Transporters Desired Catalytic Localization Gene and/or Structure Activities Sequences Deletions Phosphatases Man 5 GlcNAc 2 ⁇ -1,2- Mns1 (N-terminus, OCH1 none mannosidase S. cerevisiae ) MNN4 (murine, Och1 (N-terminus, MNN6 human, S. cerevisiae , P.
  • the gene locus encoding alpha-1,6 mannosyltransferase activity therefore is a prime target for the integration of genes encoding glycosyltransferase activity.
  • a range of other chromosomal integration sites that, based on a gene disruption event in that locus, are expected to: (1) improve the cells ability to glycosylate in a more human-like fashion, (2) improve the cells ability to secrete proteins, (3) reduce proteolysis of foreign proteins and (4) improve other characteristics of the process that facilitate purification or the fermentation process itself.
  • glycoproteins especially glycoproteins used therapeutically in humans.
  • Glycoproteins having specific glycoforms may be especially useful, for example, in the targeting of therapeutic proteins.
  • mannose-6-phosphate has been shown to direct proteins to the lysosome, which may be essential for the proper function of several enzymes related to lysosomal storage disorders such as Gaucher's, Hunter's, Hurler's, Scheie's, Fabry's and Tay-Sachs disease, to mention just a few.
  • the addition of one or more sialic acid residues to a glycan side chain may increase the lifetime of a therapeutic glycoprotein in vivo after administration.
  • host cells e.g., lower eukaryotic or mammalian
  • sialic acid may be conjugated to the protein of interest in vitro prior to administration using a sialic acid transferase and an appropriate substrate.
  • Changes in growth medium composition may be employed in addition to the expression of enzyme activities involved in human-like glycosylation to produce glycoproteins more closely resembling human forms (Weikert et al. (1999) Nature Biotechnology 17, 1116-1121; Werner et al. (1998) Arzneiffenforschung 48(8):870-880; Andersen and Goochee (1994) Cur. Opin.
  • Therapeutic proteins are typically administered by injection, orally, pulmonary, or other means.
  • suitable target glycoproteins which may be produced according to the invention include, without limitation: erythropoietin, cytokines such as interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , and granulocyte-CSF, coagulation factors such as factor VIII, factor IX, and human protein C, soluble IgE receptor ⁇ -chain, IgG, IgG fragments, IgM, interleukins, urokinase, chymase, urea trypsin inhibitor, IGF-binding protein, epidermal growth factor, growth hormone-releasing factor, annexin V fusion protein, angiostatin, vascular endothelial growth factor-2, myeloid progenitor inhibitory factor-1, osteoprotegerin, ⁇ -1-antitrypsin, Dnase II, and ⁇ -feto proteins.
  • N-acetylglucosamine residues to the GlcNAcMan 3 GlcNAc 2 structure by N-acetylglucosaminyltransferases II and III yields a so-called bisected N-glycan GlcNAc 3 Man 3 GlcNAc 2 ( FIG. 15 ).
  • This structure has been implicated in greater antibody-dependent cellular cytotoxicity (ADCC) (Umana et al. (1999) Nat. Biotechnol. 17(2):176-80). Re-engineering glycoforms of immunoglobulins expressed by mammalian cells is a tedious and cumbersome task.
  • ADCC antibody-dependent cellular cytotoxicity
  • the invention provides systems and methods for producing human-like N-glycans having bisecting N-acetylglucosamine (GlcNAc) on a trimannose or pentamannose core structure.
  • the invention provides a system and method for producing immunoglobulins having bisected N-glycans.
  • the systems and methods described herein will not suffer from previous problems, e.g., cytotoxicity associated with overexpression of GnTIII or ADCC, as the host cells of the invention are engineered and selected to be viable and preferably robust cells which produce N-glycans having substantially modified human-type glycoforms such as GlcNAc 2 Man 3 GlcNAc 2 .
  • addition of a bisecting N-acetylglucosamine in a host cell of the invention will have a negligible effect on the growth-phenotype or viability of those host cells.
  • a host cell such as a yeast strain capable of producing glycoproteins with bisecting N-glycans is engineered according to the invention, by introducing into the host cell a GnTIII activity (Example 12).
  • the host cell is transformed with a nucleic acid that encodes GnTIII (see, e.g., FIG. 24 ) or a domain thereof having enzymatic activity, optionally fused to a heterologous cell signal targeting peptide (e.g., using the libraries and associated methods of the invention.)
  • Host cells engineered to express GnTIII will produce higher antibody titers than mammalian cells are capable of. They will also produce antibodies with higher potency with respect to ADCC.
  • Antibodies produced by mammalian cell lines transfected with GnTIII have been shown to be as effective as antibodies produced by non-transfected cell-lines, but at a 10-20 fold lower concentration (Davies et al. (2001) Biotechnol. Bioeng. 74(4):288-94).
  • the invention thus provides a system and method for producing high titers of an antibody having high potency (e.g., up to several orders of magnitude more potent than what can currently be produced).
  • the system and method is safe and provides high potency antibodies at low cost in short periods of time.
  • Host cells engineered to express GnTIII according to the invention produce immunoglobulins having bisected N-glycans at rates of at least 50 mg/liter/day to at least 500 mg/liter/day.
  • each immunoglobulin (Ig) molecule (comprising bisecting GlcNAcs) is more potent than the same Ig molecule produced without bisecting GlcNAcs.
  • a 1215 bp ORF of the P. pastoris OCH1 gene encoding a putative ⁇ -1,6 mannosyltransferase was amplified from P. pastoris genomic DNA (strain X-33, Invitrogen, Carlsbad, Calif.) using the oligonucleotides 5′-ATGGCGAAGGCAGATGGCAGT-3′ (SEQ ID NO:3) and 5′-TTAGTCCTTCCAACTTCCTTC-3′ (SEQ ID NO:4) which were designed based on the P. pastoris OCH1 sequence (Japanese Patent Application Publication No. 8-336387).
  • OCH1 mutants of S. cerevisiae are temperature sensitive and are slow growers at elevated temperatures.
  • OCH1 mutants of S. cerevisiae are temperature sensitive and are slow growers at elevated temperatures.
  • One can thus identify functional homologs of OCH1 in P. pastoris by complementing an OCH1 mutant of S. cerevisiae with a P. pastoris DNA or cDNA library.
  • About 20 temperature sensitive strains were further subjected to a colony PCR screen to identify colonies with a deleted och1 gene. Several och1 deletions were obtained.
  • the linearized pBK9.1 which has 2.1 kb upstream sequence and 1.5 kb downstream sequence of OCH1 gene cassette carrying Pichia HIS4 gene, was transformed into P. pastoris BK1 [GS1 15 (his4 Invitrogen Corp., San Diego, Calif.) carrying the human IFN- ⁇ gene in the AOX1 locus] to knock out the wild-type OCH1 gene.
  • the initial screening of transformants was performed using histidine drop-out medium followed by replica plating to select the temperature sensitive colonies. Twenty out of two hundred histidine-positive colonies showed a temperature sensitive phenotype at 37° C.
  • the 20 temperature-sensitive isolates were subjected to colony PCR using primers specific to the upstream sequence of the integration site and to HIS4 ORF. Two out of twenty colonies were ochl defective and further analyzed using a Southern blot and a Western blot indicating the functional och1 disruption by the och1 knock-out construct. Genomic DNA were digested using two separate restriction enzymes BglII and ClaI to confirm the och1 knock-out and to confirm integration at the open reading frame. The Western Blot showed och1 mutants lacking a discrete band produced in the GS 115 wild type at 46.2 kDa.
  • Man 5 GlcNAc 2 an essential intermediate for complex N-glycan formation. While the production of a Man 5 GlcNAc 2 precursor is essential, it is not necessarily sufficient for the production of hybrid and complex glycans because the specific isomer of Man 5 GlcNAc 2 may or may not be a substrate for GnTI.
  • An och1 mutant of P. pastoris is engineered to express secreted human interferon- ⁇ under the control of an aox promoter.
  • a DNA library is constructed by the in-frame ligation of the catalytic domain of human mannosidase IB (an ⁇ -1,2-mannosidase) with a sub-library including sequences encoding early Golgi and ER localization peptides.
  • the DNA library is then transformed into the host organism, resulting in a genetically mixed population wherein individual transformants each express interferon- ⁇ as well as a synthetic mannosidase gene from the library. Individual transformant colonies are cultured and the production of interferon is induced by addition of methanol. Under these conditions, over 90% of the secreted protein is glycosylated interferon- ⁇ .
  • Supernatants are purified to remove salts and low-molecular weight contaminants by C 18 silica reversed-phase chromatography. Desired transformants expressing appropriately targeted, active ⁇ -1,2-mannosidase produce interferon- ⁇ including N-glycans of the structure Man 5 GlcNAc 2 , which has a reduced molecular mass compared to the interferon- ⁇ of the parent strain.
  • the purified interferon- ⁇ is analyzed by MALDI-TOF mass spectroscopy and colonies expressing the desired form of interferon- ⁇ are identified.
  • the 1215 bp open reading frame of the P. pastoris OCH1 gene as well as 2685 bp upstream and 1175 bp downstream was amplified by PCR (see also WO 02/00879), cloned into the pCR2.1-TOPO vector (Invitrogen) and designated pBK9.
  • pBK9 pCR2.1-TOPO vector
  • Plasmid pPB103 containing the Kluyveromyces lactis MNN2-2 gene which encodes a Golgi UDP-N-acetylglucosamine transporter was constructed by cloning a blunt BglII-HindIII fragment from vector pDL02 (Abeijon et al. (1996) Proc. Natl. Acad. Sci. U.S.A. 93:5963-5968) into BgllI and BamHI digested and blunt ended pBLADE-SX containing the P. pastoris ADE1 gene (Cereghino et al. (2001) Gene 263:159-169).
  • This plasmid was linearized with EcoNI and transformed into strain BK64-1 by electroporation and one strain confirmed to contain the MNN2-2 by PCR analysis was named PBP1.
  • a library of mannosidase constructs was generated, comprising in-frame fusions of the leader domains of several type I or type II membrane proteins from S. cerevisiae and P. pastoris fused with the catalytic domains of several ⁇ -1,2-mannosidase genes from human, mouse, fly, worm and yeast sources (see, e.g., WO02/00879, incorporated herein by reference).
  • This library was created in a P. pastoris HIS4 integration vector and screened by linearizing with SalI, transforming by electroporation into strain PBP1, and analyzing the glycans released from the K3 reporter protein.
  • a library of GnTI constructs was generated, comprising in-frame fusions of the same leader library with the catalytic domains of GnTI genes from human, worm, frog and fly sources (WO 02/00879).
  • This library was created in a P. pastoris ARG4 integration vector and screened by linearizing with AatII, transforming by electroporation into strain PBP2, and analyzing the glycans released from K3.
  • One active construct chosen was a chimera of the first 120 bp of the S. cerevisiae MNN9 gene fused to a deletion of the human GnTI gene, which was missing the first 154 bp.
  • a P. pastoris strain expressing this construct was named PBP-3. (See also FIG. 36 .)
  • a library of GnTII constructs was generated, which comprised in-frame fusions of the leader library with the catalytic domains of GnTII genes from human and rat sources (WO 02/00879).
  • This library was created in a P. pastoris integration vector containing the NST R gene conferring resistance to the drug nourseothricin.
  • the library plasmids were linearized with EcoRI, transformed into strain RDP27 by electroporation, and the resulting strains were screened by analysis of the released glycans from purified K3.
  • MOPS sodium cacodylate, manganese chloride, UDP-galactose and CMP-N-acetylneuraminic acid were from Sigma.
  • Trifluoroacetic acid (TFA) was from Sigma/Aldrich, Saint Louis, Mo.
  • Recombinant rat ⁇ 2,6-sialyltransferase from Spodoptera frugiperda and ⁇ 1,4-galactosyltransferase from bovine milk were from Calbiochem (San Diego, Calif.).
  • Protein N-glycosidase F, mannosidases, and oligosaccharides were from Glyko (San Rafael, Calif.).
  • DEAE ToyoPearl resin was from TosoHaas.
  • Metal chelating “HisBind” resin was from Novagen (Madison, Wis.). 96-well lysate-clearing plates were from Promega (Madison, Wis.). Protein-binding 96-well plates were from Millipore (Bedford, Mass.). Salts and buffering agents were from Sigma (St. Louis, Mo.). MALDI matrices were from Aldrich (Milwaukee, Wis.).
  • Kringle 3 was purified using a 96-well format on a Beckman BioMek 2000 sample-handling robot (Beckman/Coulter Collins Cucamonga, Calif.). Kringle 3 was purified from expression media using a C-terminal hexa-histidine tag.
  • the robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin.
  • a 150 uL ( ⁇ L) settled volume of resin is poured into the wells of a 96-well lysate-binding plate, washed with 3 volumes of water and charged with 5 volumes of 50 mM NiSO4 and washed with 3 volumes of binding buffer (5 mM imidazole, 0.5 M NaCl, 20 mM Tris-HCL pH7.9).
  • the protein expression media is diluted 3:2, media/PBS (60 mM PO4, 16 mM KCl, 822 mM NaCl pH7.4) and loaded onto the columns.
  • the columns are washed with 10 volumes of binding buffer and 6 volumes of wash buffer (30 mM imidazole, 0.5 M NaCl, 20 mM Tris-HCl pH7.9) and the protein is eluted with 6 volumes of elution buffer (1 M imidazole, 0.5 M NaCl, 20 mM Tris-HCl pH7.9).
  • the eluted glycoproteins are evaporated to dryness by lyophilyzation.
  • the glycans are released and separated from the glycoproteins by a modification of a previously reported method (Papac, et al. A. J. S. (1998) Glycobiology 8, 445-454).
  • the wells of a 96-well MultiScreen IP (Immobilon-P membrane) plate (Millipore) are wetted with 100 uL of methanol, washed with 3 ⁇ 150 uL of water and 50 uL of RCM buffer (8M urea, 360 mM Tris, 3.2 mM EDTA pH8.6), draining with gentle vacuum after each addition.
  • the dried protein samples are dissolved in 30 uL of RCM buffer and transferred to the wells containing 10 uL of RCM buffer.
  • the wells are drained and washed twice with RCM buffer.
  • the proteins are reduced by addition of 60 uL of 0.1 M DTT in RCM buffer for 1 hr at 37° C.
  • the wells are washed three times with 300 uL of water and carboxymethylated by addition of 60 uL of 0.1 M iodoacetic acid for 30min in the dark at room temperature.
  • the wells are again washed three times with water and the membranes blocked by the addition of 100 uL of 1% PVP 360 in water for 1 hr at room temperature.
  • the wells are drained and washed three times with 300 uL of water and deglycosylated by the addition of 30 uL of 10 mM NH 4 HCO 3 pH 8.3 containing one milliunit of N-glycanase (Glyko). After 16 hours at 37° C., the solution containing the glycans was removed by centrifugation and evaporated to dryness.
  • glycans were determined using a Voyager DE PRO linear MALDI-TOF (Applied Biosciences) mass spectrometer using delayed extraction. The dried glycans from each well were dissolved in 15 uL of water and 0.5 uL spotted on stainless steel sample plates and mixed with 0.5 uL of S-DHB matrix (9 mg/mL of dihydroxybenzoic acid, 1 mg/mL of 5-methoxysalicilic acid in 1:1 water/acetonitrile 0.1% TFA) and allowed to dry.
  • S-DHB matrix 9 mg/mL of dihydroxybenzoic acid, 1 mg/mL of 5-methoxysalicilic acid in 1:1 water/acetonitrile 0.1% TFA
  • Ions were generated by irradiation with a pulsed nitrogen laser (337 nm) with a 4 ns pulse time.
  • the instrument was operated in the delayed extraction mode with a 125 ns delay and an accelerating voltage of 20 kV.
  • the grid voltage was 93.00%
  • guide wire voltage was 0.10%
  • the internal pressure was less than 5 ⁇ 10-7 torr
  • the low mass gate was 875 Da.
  • Spectra were generated from the sum of 100-200 laser pulses and acquired with a 2 GHz digitizer. Man 5 GlcNAc 2 oligosaccharide was used as an external molecular weight standard. All spectra were generated with the instrument in the positive ion mode. The estimated mass accuracy of the spectra was 0.5%.
  • An och1 mutant of P. pastoris was engineered to express and secrete proteins such as the kringle 3 domain of human plasminogen (K3) under the control of the inducible AOXI promoter.
  • the Kringle 3 domain of human plasminogen (K3) was used as a model protein.
  • a DNA fragment encoding the K3 was amplified using Pfu turbo polymerase (Strategene, La Jolla, Calif.) and cloned into EcoRI and XbaI sites of pPICZaA (Invitrogen, Carlsbad, Calif.), resulting in a C-terminal 6- His tag.
  • Pro 46 was replaced with Ser 46 using site-directed mutagenesis.
  • the resulting plasmid was designated pBK64.
  • the correct sequence of the PCR construct was confirmed by DNA sequencing.
  • a combinatorial DNA library was constructed by the in-frame ligation of murine ⁇ -1,2-mannosidase IB (Genbank AN 6678787) and IA (Genbank AN 6754619) catalytic domains with a sub-library including sequences encoding Cop II vesicle, ER, and early Golgi localization peptides according to Table 6.
  • the combined DNA library was used to generate individual fusion constructs, which were then transformed into the K3 expressing host organism, resulting in a genetically mixed population wherein individual transformants each express K3 as well as a localization signal/mannosidase fusion gene from the library. Individual transform ants were cultured and the production of K3 was induced by transfer to a methanol containing medium.
  • the K3 reporter protein was purified from the supernatant to remove salts and low-molecular weight contaminants by Ni-affinity chromatography. Following affinity purification, the protein was desalted by size exclusion chromatography on a Sephadex G10 resin (Sigma, St. Louis, Mo.) and either directly subjected to MALDI-TOF analysis described below or the N-glycans were removed by PNGase digestion as described below (Release of N-glycans) and subjected to MALDI-TOF analysis Miele et al. (1997) Biotechnol. Appl. Biochem. 25:151-157.
  • Targeting peptides were selected from MNS I (SwissProt P32906) in S. cerevisiae (long, medium and short) (see supra Nucleic Acid Libraries; Combinatorial DNA Library of Fusion Constructs) and SEC12 (SwissProt P11655) in S. cerevisiae (988-1140 nucleotides: short) and (988-1296: medium). Although majority of the targeting peptide sequences were N-terminal deletions, some targeting peptide sequences, such as SEC12 were C-terminal deletions.
  • Catalytic domains used in this experiment were selected from mouse mannosidase 1A with a 187 amino acid N-terminal deletion; and mouse mannosidase 1B with a 58, 99 and 170 amino acid deletion.
  • the number of (+)s, as used herein, indicates the relative levels of Man 5 GlcNAc 2 production.
  • the notation ( ⁇ ) indicates no apparent production of Man 5 GlcNAc 2 .
  • the notation (+) indicates less than 10% production of Man 5 GlcNAc 2
  • the notation (++) indicates about 10-20% production of Man 5 GlcNAc 2 .
  • the notation with (+++) indicates about 20-40% production of Man 5 GlcNAc 2 .
  • the notation with (++++) indicates about 50% production of Man 5 GlcNAc 2 .
  • the notation with (+++++) indicates greater than 50% production of Man 5 GlcNAC 2 .
  • Table 9 shows relative amount of Man 5 GlcNAc 2 on secreted K3.
  • Six hundred and eight (608) different strains of P. pastoris , ⁇ och1 were generated by transforming them with a single construct of a combinatorial genetic library that was generated by fusing nineteen (19) ⁇ -1,2 mannosidase catalytic domains to thirty-two (32) fungal ER, and cis-Golgi leaders.
  • Table 7 shows two constructs pFB8 and pGC5, among others, displaying Man 5 GlcNAc 2 .
  • Table 8 shows a more preferred construct, pBC18-5, a S. cerevisiae VAN1 (S) targeting peptide sequence (from SwissProt 23642) ligated in-frame to a C. elegans mannosidase IB (Genbank AN CAA98114) 80 amino acid N-terminal deletion ( Saccharomyces Van1 (s)/ C. elegans mannosidase IB ⁇ 80).
  • This fusion construct also produces a predominant Man 5 GlcNAc 2 structure, as shown in FIG. 5E . This construct was shown to produce greater than 50% Man 5 GlcNAc 2 (+++++).
  • Generating a combinatorial DNA library of ⁇ -1,2-mannosidase catalytic domains fused to targeting peptides required the amplification of mannosidase domains with varying lengths of N-terminal deletions from a number of organisms.
  • ORFs open reading frames
  • ⁇ -1,2-mannosidases were PCR amplified from either cDNA or genomic DNA obtained from the following sources: Homo sapiens, Mus musculus, Drosophila melanogaster, Caenorhabditis elegans, Aspergillus nidulans and Penicillium citrinum.
  • DNA was incubated in the presence of oligonucleotide primers specific for the desired mannosidase sequence in addition to reagents required to perform the PCR reaction.
  • oligonucleotide primers specific for the desired mannosidase sequence for example, to amplify the ORF of the M. musculus ⁇ -1,2-mannosidase IA, the 5′-primer ATGCCCGTGGGGGGCCTGTTGCCGCTCTTCAGTAGC (SEQ ID NO:52) and the 3′-primer TCATTTCTCTTTGCCATCAATTTCCTTCTTCTGTTCACGG (SEQ ID NO:53) were incubated in the presence of Pfu DNA polymerase (Stratagene, La Jolla, Calif.) and amplified under the conditions recommended by Stratagene using the cycling parameters: 94° C.
  • each mannosidase was used as the template in a subsequent round of PCR reactions wherein the annealing position of the 5′-primer was specific to the 5′-terminus of the desired truncation and the 3′-primer remained specific for the original 3′-terminus of the ORF.
  • pJN347 FIG. 2C
  • AscI and Pacl restriction sites were engineered onto each truncation product, at the 5′- and 3′-termini respectively.
  • the number and position of the N-terminal truncations generated for each mannosidase ORF depended on the position of the transmembrane (TM) region in relation to the catalytic domain (CD). For instance, if the stem region located between the TM and CD was less than 150 bp, then only one truncation for that protein was generated. If, however, the stem region was longer than 150 bp then either one or two more truncations were generated depending on the length of the stem region.
  • FIG. 3 illustrates the ORF of the M musculus ⁇ -1,2-mannosidase IA with the predicted transmembrane and catalytic domains being highlighted in bold. Based on this structure, three 5′-primers were designed (annealing positions underlined in FIG. 3 ) to generate the ⁇ 65-, ⁇ 105- and ⁇ 187-N-terminal deletions.
  • the 5′-primer used was 5′-GGCGCGCCGACTCCTCCAAGCTGCTCAGCGGGGTCCTGTTCCAC-3′ (SEQ ID NO:54) (with the AscI restriction site highlighted in bold) in conjunction with the 3′-primer 5′-CCTTAATTAATCATTTCTCTTTGCCATCAATTTCCTTCTTCTGTTCACGG-3′ (SEQ ID NO:55) (with the PacI restriction site highlighted in bold). Both of these primers were used to amplify a 1561 bp fragment under the conditions outlined above for amplifying the full length M. musculus mannosidase 1A ORF.
  • the truncated product was also incubated with Taq DNA polymerase, ligated into pCR2.1-TOPO (Invitrogen, Carlsbad, Calif.), transformed into TOP10 and ABI sequenced. After having amplified and confirmed the sequence of the truncated mannosidase fragment, the resulting plasmid, pCR2.1- ⁇ 65 mMannIA, was digested with AscI and PacI in New England Biolabs buffer #4 (Beverly, Mass.) for 16 h at 37° C. In parallel, the pJN347 ( FIG. 2C ) was digested with the same enzymes and incubated as described above.
  • both the pJN347 ( FIG. 2C ) back-bone and the truncated catalytic domain were gel extracted and ligated using the Quick Ligation Kit (New England Biolabs, Beverly, Mass.), as recommended by the manufacturers, and transformed into chemically competent DH5 ⁇ cells (Invitrogen, Carlsbad, Calif.). Colony PCR was used to confirm the generation of the pJN347-mouse Mannosidase IA ⁇ 65 construct.
  • the remaining step in generating the targeting peptide/catalytic domain library was to clone in-frame the targeting peptide sequences ( FIG. 2 ).
  • Both the pJN347-mannosidase constructs ( FIG. 2D ) and the pCR2.1TOPO-targeting peptide constructs ( FIG. 2B ) such as were incubated overnight at 37° C. in New England Biolabs buffer #4 in the presence of the restriction enzymes NotI and AscI.
  • both the pJN347-mannosidase back-bone and the targeting peptide regions were gel-extracted and ligated using the Quick Ligation Kit (New England Biolabs, Beverly, Mass.), as recommended by the manufacturers, and transformed into chemically competent DH5 ⁇ cells (Invitrogen, Carlsbad, Calif.). Subsequently, the pJN347-targeting peptide/mannosidase constructs were ABI sequenced to confirm that the generated fusions were in-frame.
  • the estimated size of the final targeting peptide/alpha-1,2-mannosidase library contains over 1300 constructs generated by the approach described above.
  • FIG. 2 illustrates construction of the combinatorial DNA library.
  • the first step in plasmid construction involved creating a set of universal plasmids containing DNA regions of the KEX1 gene of P. pastoris (Boehm et al. Yeast 1999 May;15(7):563-72) as space holders for the 5′ and 3′ regions of the genes to be knocked out.
  • the plasmids also contained the S. cerevisiae Ura-blaster (Alani et al. (1987) Genetics 116:541-545) as a space holder for the auxotrophic markers, and an expression cassette with a multiple cloning site for insertion of a foreign gene.
  • pastoris KEXI-5′ region was amplified by PCR using primers GGCGAGCTCGGCCTACCCGGCCAAGGCTGAGATCATTTGTCCAGCTTCA GA (SEQ ID NO:56) and GCCCACGTCGACGGATCCGTTTAAACATCGATTGGAGAGGCTGACACC GCTACTA (SEQ ID NO:57) and P. pastoris genomic DNA as a template and cloned into the SacI, SalI sites of pUC19 (New England Biolabs, Beverly, Mass.).
  • the resulting plasmid was cut with BamHI and SalI, and a 0.8-kb fragment of the KEX1-3′ region that had been amplified using primers CGGGATCCACTAGTATTTAAATCATATGTGCGAGTGTACAACTCTTCCC ACATGG (SEQ ID NO:58) and GGACGCGTCGACGGCCTACCCGGCCGTACGAGGAATTTCTCGG ATGACTCTTTTC (SEQ ID NO:59) was cloned into the open sites creating pJN262.
  • This plasmid was cut with BamHI and the 3.8-kb BamHI, BglII fragment of pNKY51 (Alani et al. (1987) Genetics 116:541-545) was inserted in both possible orientations resulting in plasmids pJN263 ( FIG. 4A ) and pJN284 ( FIG. 4B ).
  • An expression cassette was created with NotI and PacI as cloning sites.
  • the GAPDH promoter of P. pastoris was amplified using primers CGGGATCCCTCGAGAGATCTTTTTTGTAGAAATGTCTTGGTGCCT (SEQ ID NO:60) and GGACATGCATGCACTAGTGCGGCCGCCACGTGATAGTTGTTCA ATTGATTGAAATAGGGACAA (SEQ ID NO:61) and plasmid pGAPZ-A (Invitrogen) as template and cloned into the BamHI, SphI sites of pUC 19 (New England Biolabs, Beverly, Mass.) ( FIG. 4B ).
  • the resulting plasmid was cut with SpeI and SphI and the CYC1 transcriptional terminator region (“TT”) that had been amplified using primers CCTTGCTAGCTTAATTAACCGCGGCACGTCCGACGGCGGCCCA CGGGTCCCA (SEQ ID NO:62) and GGACATGCATGCGGATCCCTTAAGAGCCGGCAGCTTGCAAATT AAAGCCTTCGAGCGTCCC (SEQ ID NO:63) and plasmid pPICZ-A (Invitrogen) as a template was cloned into the open sites creating pJN261 ( FIG. 4B ).
  • TT transcriptional terminator region
  • a knockout plasmid for the P. pastoris OCH1 gene was created by digesting pJN263 with SalI and SpeI and a 2.9-kb DNA fragment of the OCH1-5′ region, which had been amplified using the primers GAACCACGTCGACGGCCATTGCGGCCAAAACCTTTTTTCCTATT CAAACACAAGGCATTGC (SEQ ID NO:64) and CTCCAATACTAGTCGAAGATTATCTTCTACGGTGCCTGGACTC (SEQ ID NO:65) and P. pastoris genomic DNA as a template, was cloned into the open sites ( FIG. 4C ).
  • the resulting plasmid was cut with EcoRI and PmeI and a 1.0-kb DNA fragment of the OCH1-3′ region that had been generated using the primers TGGAAGGTTTAAACAAAGCTAGAGTAAAATAGATATAGCGAG ATTAGAGAATG (SEQ ID NO:66) and AAGAATTCGGCTGGAAGGCCTTGTACCTTGATGTAGTTCCCGTT TTCATC (SEQ ID NO:67) was inserted to generate pJN298 ( FIG. 4C ).
  • the BamHI expression cassette of pJN261 FIG. 4B
  • the P. pastoris Ura3-blaster cassette was constructed using a similar strategy as described in Lu et al. (1998) Appl. Microbiol. Biotechnol. 49:141-146.
  • a 2.0-kb PstI, Spel fragment of P. pastoris URA3 was inserted into the PstI, XbaI sites of pUC19 (New England Biolabs, Beverly, Mass.) to create pJN306 ( FIG. 4D ).
  • pJN306 FIG. 4D
  • a 0.7-kb SacI, PvuII DNA fragment of the lacZ open reading frame was cloned into the Sacl, SmaI sites to yield pJN308 ( FIG. 4D ).
  • Following digestion of pJN308 FIG.
  • a HIS4 marked expression plasmid was created by cutting pJN261 ( FIG. 4F ) with EcoICRI ( FIG. 4F ).
  • a 2.7 kb fragment of the Pichia pastoris HIS4 gene that had been amplified using the primers GCCCAAGCCGGCCTTAAGGGATCTCCTGATGACTGACTCACTGATAATA AAAATACGG (SEQ ID NO:68) and GGGCGCGTATTTAAATACTAGTGGATCTATCGAATCTAAATGTAAGTTA AAATCTCTAA (SEQ ID NO:69) cut with NgoMIV and SwaI and then blunt-ended using T4 DNA polymerase, was then ligated into the open site.
  • This plasmid was named pJN337 ( FIG.
  • pJN337 was cut with NotI and PacI and the two oligonucleotides GGCCGCCTGCAGATTTAAATGAATTCGGCGCGCCTTAAT (SEQ ID NO:70) and TAAGGCGCGCCGAATTCATTTAAATCTGCAGGGC (SEQ ID NO:71) that had been annealed in vitro were ligated into the open sites, creating pJN347 ( FIG. 4F ).
  • pJN329 was digested with SfiI and used to transform P. pastoris strain JC308 (Cereghino et al. (2001) Gene 263:159-169) by electroporation. Following transformation, the URA dropout plates were incubated at room temperature for 10 days. One thousand (1000) colonies were picked and restreaked. All 000 clones were then streaked onto 2 sets of URA dropout plates. One set was incubated at room temperature, whereas the second set was incubated at 37° C.
  • Positive transformants screened by colony PCR confirming integration of the mannosidase construct into the P. pastoris genome were subsequently grown at room temperature in 50 ml BMGY buffered methanol-complex medium consisting of 1% yeast extract, 2% peptone, 100 mM potassium phosphate buffer, pH 6.0, 1.34% yeast nitrogen base, 4 ⁇ 10 ⁇ 5 % biotin, and 1% glycerol as a growth medium) until OD 600nm 2-6 at which point they were washed with 10 ml BMMY (buffered methanol-complex medium consisting of I % yeast extract, 2% peptone, 100 mM potassium phosphate buffer, pH 6.0, 1.34% yeast nitrogen base, 4 ⁇ 10 ⁇ 5 % biotin, and 1.5% methanol as a growth medium) media prior to induction of the reporter protein for 24 hours at room temperature in 5 ml BMMY.
  • BMMY buffered methanol-complex medium consisting of 1% yeast
  • FIGS. 5 and 6 show expression of constructs generated from the combinatorial DNA library which show significant mannosidase activity in P. pastoris .
  • Expression of pGC5 Saccharomyces MNS1 (m)/mouse mannosidase IB ⁇ 99) ( FIGS. 5D and 6E ) produced a protein which has approximately 30% of all glycans trimmed to Man 5 GlcNAc 2
  • expression of pFB8 Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187
  • FIG. 6F produced approximately 50% Man 5 GlcNAc 2
  • expression of pBC18-5 Saccharomyces VAN1 (s)/ C. elegans mannosidase IB ⁇ 80
  • FIG. 5E produced 70% Man 5 GlcNAc 2 .
  • P. pastoris cells comprising plasmid pFB8 ( Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187) were grown at 30° C. in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for mannosidase assays and the remainder was used for the recovery of secreted soluble K3.
  • plasmid pFB8 Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187
  • FIG. 7A the supernatant from the above aliquot ( FIG. 7B ), and BMMY containing 10 ng of 75 mU/mL of ⁇ -1,2-mannosidase from Trichoderma reesei (obtained from Contreras et al., WO 02/00856 A2) ( FIG. 7C ). After incubation for 24 hours at room temperature, samples were analyzed by amino silica HPLC to determine the extent of mannosidase trimming.
  • P. pastoris cells comprising plasmid pGC5 ( Saccharomyces MNS1 (m)/mouse mannosidase IB ⁇ 99) were similarly grown and assayed. Cells were grown at room temperature in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for mannosidase assays and the remainder was used for the recovery of secreted soluble K3.
  • plasmid pGC5 Saccharomyces MNS1 (m)/mouse mannosidase IB ⁇ 99
  • FIG. 5D N-glycan analysis of the K3 derived glycans is shown in FIG. 5D .
  • the earlier removed aliquot of the supernatant was further tested for the presence of secreted mannosidase activity as shown in FIG. 8B .
  • a commercially available standard of Man9-2-AB (Glyko, Novato, Calif.) were added to:BMMY ( FIG. 8A ), supernatant from the above aliquot ( FIG.
  • FIG. 8C BMMY containing 10 ng of 75 mU/mL of ⁇ -1,2-mannosidase from Trichoderma reesei (obtained from Contreras et al., WO 02/00856 A2) ( FIG. 8C ). After incubation for 24 hours at room temperature, samples were analyzed by amino silica HPLC to determine the extent of mannosidase trimming.
  • Man9-2-AB was used as a substrate and it is evident that after 24 hours of incubation, mannosidase activity was virtually absent in the supernatant of the pFB8 ( Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187) strain digest ( FIG. 7B ) and pGC5 ( Saccharomyces MNS1 (m)/mouse mannosidase IB ⁇ 99) strain digest ( FIG. 8B ) whereas the positive control (purified ⁇ -1,2-mannosidase from T. reesei obtained from Contreras) leads to complete conversion of Man 9 GlcNAc 2 to Man 5 GlcNAc 2 under the same conditions, as shown in FIGS.
  • FIG. 9 further substantiates localization and activity of the mannosidase enzyme.
  • P. pastoris comprising pBC18-5 Saccharomyces VAN1 (s)/ C. elegans mannosidase IB ⁇ 80
  • pBC18-5 Saccharomyces VAN1 (s)/ C. elegans mannosidase IB ⁇ 80
  • FIG. 9A supernatant from the above aliquot pBC18-5 ( Saccharomyces VAN1 (s)/ C. elegans mannosidase IB ⁇ 80) ( FIG. 9B ), and BMMY containing media from a different fusion construct pDD28-3 ( Saccharomyces MNN10 (m) (from SwissProt 50108)/ H. sapiens mannosidase IB ⁇ 99) ( FIG. 9C ). After incubation for 24 hours at room temperature,. samples were analyzed by amino silica HPLC to determine the extent of mannosidase trimming.
  • FIG. 9C After incubation for 24 hours at room temperature,. samples were analyzed by amino silica HPLC to determine the extent of mannosidase trimming.
  • FIG. 9B demonstrates intracellular mannosidase activity in comparison to a fusion construct pDD28-3 ( Saccharomyces MNN10 (m) H. sapiens mannosidase IB ⁇ 99) exhibiting a negative result ( FIG. 9C ).
  • P. pastoris cells comprising plasmid pBB27-2 ( Saccharomyces MANN10 (s) (from SwissProt 50108)/ C. elegans mannosidase IB ⁇ 31) were grown at room temperature in BMGY to an OD600 of about 17. About 80 ⁇ L of these cells were inoculated into 600 ⁇ L BMGY and were grown overnight. Subsequently, cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant (pH 6.43).
  • Saccharomyces MANN10 Saccharomyces MANN10 (s) (from SwissProt 50108)/ C. elegans mannosidase IB ⁇ 31
  • GlcNAc Transferase I activity is required for the maturation of complex and hybrid N-glycans (U.S. Pat. No. 5,834,251).
  • Man 5 GlcNAc 2 may only be trimmed by mannosidase II, a necessary step in the formation of human glycoforms, after the addition of N-acetylglucosamine to the terminal ⁇ -1,3 mannose residue of the trimannose stem by GlcNAc Transferase I (Schachter, 1991 Glycobiology 1(5):453-461). Accordingly, a combinatorial DNA library was prepared including DNA fragments encoding suitably targeted catalytic domains of GlcNAc Transferase I genes from C.
  • elegans and Homo sapiens and localization sequences from GLS, MNS, SEC, MNN9, VAN1, ANP1, HOC1, MNN10, MNN11, MNT1, KTR1, KTR2, MNN2, MNN5, YUR1, MNN1, and MNN6 from S. cerevisiae and P. pastoris putative ⁇ -1,2-mannosyltransferases based on the homology from S. cerevisiae: D2, D9 and J3, which are KTR homologs.
  • Table 10 includes but does not limit targeting peptide sequences such as SEC and OCH1, from P. pastoris and K.
  • lactis GnTI (See Table 6 and Table 10) TABLE 10 A representative combinatorial library of targeting peptide sequences/ catalytic domain for UDP-N-Acetylglucosaminyl Transferase I (GnTI) Targeting peptide OCHI(s) OCHI(m) OCHI(l) MNN9(s) MNN9(m) Catalytic Human, GnTI, ⁇ 38 PB105 PB106 PB107 PB104 N/A Domain Human, GnTI, ⁇ 86 NB12 NB13 NB14 NB15 NB C. elegans , GnTI, ⁇ 88 OA12 OA13 OA14 OA15 OA16 C.
  • Targeting peptide sequences were selected from OCH1 in P. pastoris (long, medium and short) (see Example 4) and MNN9 (SwissProt P39107) in S . cerevisiae (short and medium).
  • Catalytic domains were selected from human GnTI with a 38 and 86 amino acid N-terminal deletion, C. elegans (gly-12) GnTI with a 35 and 63 amino acid deletion as well as C. elegans (gly-14) GnTI with a 88 amino acid N-terminal deletion and X. leavis GnTI with a 33 and 103 amino acid N-terminal deletion, respectively.
  • a portion of the gene encoding human N-acetylglucosaminyl Transferase I was amplified by PCR using oligonucleotides 5′-TGGCAGGCGCGCCTCAGTCAGCGCTCTCG-3′ (SEQ ID NO:72) and 5′-AGGTTAATTA AGTGCTAATTCCAGCTAGG-3′ (SEQ ID NO:73) and vector pHG4.5 (ATCC# 79003) as template.
  • the resulting PCR product was cloned into pCR2.1-TOPO and the correct sequence was confirmed.
  • the truncated GnTI was inserted into plasmid pJN346 to create pNA.
  • the 120 bp insert was ligated into pNA to generate an in-frame fusion of the MNN9 transmembrane domain with the GnTI, creating pNA 15.
  • the host organism is a strain of P. pastoris that is deficient in hypermannosylation (e.g. an och1 mutant), provides the substrate UDP-GlcNAc in the Golgi and/or ER (i.e., contains a functional UDP-GlcNAc transporter), and provides N-glycans of the structure Man 5 GlcNAc 2 in the Golgi and/or ER (e.g. P. pastoris pFB8 ( Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187) from above).
  • P. pastoris pFB8 Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187
  • pastoris pFB8 was transformed with pPB103 containing the Kluyveromyces lactis MAN2-2 gene (Genbank AN AF 106080) (encoding UDP-GlcNAc transporter) cloned into BamHI and BglII site of pBLADE-SX plasmid (Cereghino et al. (2001) Gene 263:159-169). Then the aforementioned combinatorial DNA library encoding a combination of exogenous or endogenous GnTI/localization genes was transformed and colonies were selected and analyzed for the presence of the GnTI construct by colony PCR. Our transformation and integration efficiency was generally above 80% and PCR screening can be omitted once robust transformation parameters have been established.
  • K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot.
  • the robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin.
  • Another screening method may be performed using a specific terminal GlcNAc binding antibody, or a lectin such as the GSII lectin from Griffonia simplificolia, which binds terminal GlcNAc (EY Laboratories, San Mateo, Calif.). These screens can be automated by using lectins or antibodies that have been modified with fluorescent labels such as FITC or analyzed by MALDI-TOF.
  • Secreted K3 can be purified by Ni-affinity chromatography, quantified and equal amounts of protein can be bound to a high protein binding 96-well plate. After blocking with BSA, plates can be probed with a GSII-FACS lectin and screened for maximum fluorescent response.
  • a preferred method of detecting the above glycosylated proteins involves the screening by MALDI-TOF mass spectrometry following the affinity purification of secreted K3 from the supernatant of 96-well cultured transformants. Transformed colonies were picked and grown to an OD600 of 10 in a 2 ml, 96-well plate in BMGY at 30° C.
  • the methods of the invention yield strains of P. pastoris that produce GlcNAcMan 5 GlcNAc 2 in high yield, as shown in FIG. 10B .
  • At least 60% of the N-glycans are GlcNAcMan 5 GlcNAc 2 .
  • results presented herein show that addition of the UDP-GlcNAc transporter along with GnTI activity produces a predominant GlcNAcMan 5 GlcNAc 2 structure, which is confirmed by the peak at 1457 (m/z) ( FIG. 10B ).
  • the P. pastoris strain expressing K3, ( ⁇ och1, arg-, ade-, his-) was transformed successively with the following vectors.
  • pFB8 Saccharomyces SEC12 (m)/mouse mannosidase IA ⁇ 187
  • pPB103 containing Kluyveromyces lactis MNN2-2 gene (Genbank AN AF 106080) (encoding UDP-GlcNAc transporter) cloned into pBLADE-SX plasmid (Cereghino et al. (2001) Gene 263:159-169) digested with BamHI and BgIII enzymes was transformed in the P. pastoris strain.
  • pPB104 containing Saccharomyces MNN9(s)/human GnTI ⁇ 38 encoding gene cloned as NotI-PacI fragment into pJN336 was transformed into the P. pastoris strain.
  • the OCH1 gene of the budding yeast S. cerevisiae encodes a 1,6-mannosyltransferase that is responsible for the first Golgi localized mannose addition to the Man 8 GlcNAc 2 N-glycan structure on secreted proteins (Nakanishi-Shindo et al. (1993) J. Biol. Chem.; 268(35):26338-45).
  • This mannose transfer is generally recognized as the key initial step in the fungal specific polymannosylation of N-glycan structures (Nakanishi-Shindo et al. (1993) J. Biol. Chem. 268(35):26338-26345; Nakayama et al.
  • the Och1p sequence from S. cerevisiae was aligned with known homologs from Candida albicans (Genbank accession # AAL49987), and P. pastoris along with the Hoc1 proteins of S. cerevisiae (Neiman et al (1997) Genetics 145(3):637-45 and K. lactis (PENDANT EST database) which are related but distinct mannosyltransferases. Regions of high homology that were in common among Och1p homologs but distinct from the Hoc1p homologs were used to design pairs of degenerate primers that were directed against genomic DNA from the K. lactis strain MG1/2 (Bianchi et al (1987) Current Genetics 12:185-192).
  • the 302 bp PCR product was used to probe a Southern blot of genomic DNA from K. lactis strain (MG1/2) with high stringency (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989). Hybridization was observed in a pattern consistent with a single gene indicating that this 302 bp segment corresponds to a portion of the K. lactis genome and K. lactis (KiOCH1) contains a single copy of the gene. To clone the entire KlOCH1 gene, the Southern blot was used to map the genomic locus.
  • a 5.2 kb BamHI/PstI fragment was cloned by digesting genomic DNA and ligating those fragments in the range of 5.2 kb into pUC 19 (New England Biolabs, Beverly, Mass.) to create a K. lactis subgenomic library.
  • This subgenomic library was transformed into E. coli and several hundred clones were tested by colony PCR using RCD 33/34.
  • the 5.2 kb clone containing the predicted KiOCH1 gene was sequenced and an open reading frame of 1362 bp encoding a predicted protein that is 46.5% identical to the S. cerevisiae OCH1 gene.
  • FIG. 12A shows a MALDI-TOF analysis of a wild type K. lactis strain producing N-glycans of Man 8 GlcNAc 2 [c] and higher.
  • MANN1 is the structural gene for the Golgi ⁇ -1,3-mannosyltransferase.
  • the product of MNN1 is a 762-amino acid type II membrane protein (Yip et al. (1994) Proc Natl Acad Sci USA. 91(7):2723-7).
  • Both N-linked and O-linked oligosaccharides isolated from mnn1 mutants lack ⁇ -1,3-mannose linkages (Raschke et al. (1973) J. Biol Chem. 248(13):4660-66).
  • the Mnn1p sequence from S. cerevisiae was used to search the K. lactis translated genomic sequences (PEDANT).
  • PEDANT K. lactis translated genomic sequences
  • An internal segment of this sequence was subsequently PCR amplified with primers KMN1 (TGCCATCTTTTAGGTCCAGGCCCGTTC) (SEQ ID NO:76) and KMN2 (GATCCCACGACGCATCGTATTTCTTTC), (SEQ ID NO:77) and used to probe a Southern blot of genomic DNA from K. lactis strain (MG 1/2).
  • This disruption allele was transformed into a strain of K. lactis by electroporation and nourseothricin resistant transformants were selected and PCR amplified for homologous insertion of the disruption allele. Strains that reveal a mutant PCR pattern may be subjected to N-glycan carbohydrate analysis of a known reporter gene.
  • FIG. 12B depicts the N-glycans from the K. lactis och1 mnn1 deletion strain observed following PNGase digestion the MALDI-TOF as described herein.
  • the predominant peak at 1908 (m/z) indicated as [d] is consistent with the mass of Man 9 GlcNAc 2 .
  • yeast expression systems can be obtained from sources such as the American Type Culture Collection, Rockville, Md. Vectors are commercially available from a variety of sources.
  • Degenerate primers were generated based on an alignment of Alg3 protein sequences from S. cerevisiae, H. sapiens, and D. melanogaster and were used to amplify an 83 bp product from P. pastoris genomic DNA: (SEQ ID NO:78) 5′-GGTGTTTTGTTTTCTAGATCTTTGCAYTAYCARTT-3′ and (SEQ ID NO:79) 5′-AGAATTTGGTGGGTAAGAATTCCARCACCAYTCRTG-3′.
  • the resulting PCR product was cloned into the pCR2.1 vector (Invitrogen, Carlsbad, Calif.) and seqence analysis revealed homology to known ALG3/RHK1/NOT56 homologs (Genbank NC — 001134.2, AF309689, NC — 003424.1). Subsequently, 1929 bp upstream and 2738 bp downstream of the initial PCR product were amplified from a P.
  • the resulting fragments were cloned into the pCR2.1-TOPO vector (Invitrogen) and sequenced. From this sequence, a 1395 bp ORF was identified that encodes a protein with 35% identity and 53% similarity to the S. cerevisiae ALG3 gene (using BLAST programs). The gene was named PpALG3.
  • PpALG3 was used to create a set of primers to generate a deletion construct of the PpALG3 gene by PCR overlap (Davidson et al (2002) Microbiol. 148(Pt 8):2607-15).
  • Primers below were used to amplify 1 kb regions 5′ and 3′ of the PpALG3 ORF and the KAN R gene, respectively: RCD142 (5′-CCACATCATCCGTGCTACATATAG-3′), (SEQ ID NO:82) RCD144 (5′-ACGAGGCAAGCTAAACAGATCTCGAAGTATCGAGGGTTATCCAG-3′), (SEQ ID NO:83) RCD145 (5′-CCATCCAGTGTCGAAAACGAGCCAATGGTTCATGTCTATAAATC-3′), (SEQ ID NO:84) RCD147 (5′-AGCCTCAGCGCCAACAAGCGATGG-3′), (SEQ ID NO:85) RCD143 (5′-CTGGATAACCCTCGATACTTCGAGATCTGTTTAGCTT
  • primers RCD142 and RCD147 were used to overlap the three resulting PCR products into a single 3.6 kb alg3::KAN R deletion allele. Identification, cloning and deletion of the ALG3 gene in K. lactis.
  • ALG3p sequences from S. cerevisiae, Drosophila melanogaster, Homo sapiens etc were aligned with K. lactis sequences (PENDANT EST database). Regions of high homology that were in common homologs but distinct in exact sequence from the homologs were used to create pairs of degenerate primers that were directed against genomic DNA from the K. lactis strain MG 1/2 (Bianchi et al, 1987).
  • the PCR product was used to probe a Southern blot of genomic DNA from K. lactis strain (MG1/2) with high stringency (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989). Hybridization was observed in a pattern consistent with a single gene.
  • This Southern blot was used to map the genomic loci. Genomic fragments were cloned by digesting genomic DNA and ligating those fragments in the appropriate size-range into pUC 19 to create a K. lactis subgenomic library. This subgenomic library was transformed into E. coli and several hundred clones were tested by colony PCR, using primers KAL-1 and KAL-2. The clones containing the predicted KiALG3 and KlALG61 genes were sequenced and open reading frames identified.
  • a P. pastoris alg3::KAN R deletion construct was generated as described in Example 10. Approximately 5 ⁇ g of the resulting PCR product was transformed into strain PBP-3 (see Example 3), and colonies were selected on YPD medium containing 200 ⁇ g/ml G418. One strain out of 20 screened by PCR was confirmed to contain the correct integration of the alg3::KAN R mutant allele and lack the wild-type allele. This strain was named RDP27 ( FIG. 36 ).
  • a library of GnTII constructs was then generated, which was comprised of in-frame fusions of the leader library with the catalytic domains of GnTII genes from human and rat sources (WO 02/00879).
  • This library was created in a P. pastoris integration vector containing the NST R gene conferring resistance to the drug nourseothricin.
  • the library plasmids were linearized with EcoRI, transformed into strain RDP27 by electroporation, and the resulting strains were screened by analysis of the released glycans from purified K3.
  • a P. pastoris strain expressing the rat GnTII fused in-frame to the S. cerevisiae MNN9 (s) construct was named PBP6-5 ( FIG. 36 ).
  • GnTI expression vector containing a human GnTI gene fused with the N-terminal part of S. cerevisiae MNN9 gene is described in Choi et al. (2003) Proc Natl Acad Sci U S A. 100(9):5022-27. In a similar fashion, the rat GnTII gene was cloned.
  • the rat GnTII gene (GenBank accession number U21662) was PCR amplified using Takara EX TaqTM polymerase (Panvera) from rat liver cDNA library (Clontech) with RAT1 (5′-TTCCTCACTGCAGTCTTCTATAACT-3′) (SEQ ID NO:90) and RAT2 (5′-TGGAGACCATGAGGTTCCGCATCTAC-3′) (SEQ ID NO:91) primers.
  • the PCR product was then cloned into pCR2.1-TOPO vector (Invitrogen) and sequenced.
  • the AscI-Pacl fragment of GnTII encoding amino-acids 88-443
  • Pfu Turbo polymerase (Stratagene) and primers, RAT44 (5′-TTGGCGCGCCTCCCTAGTGTACCAGTTGAACTTTG-3′) (SEQ ID NO:92) and RAT11 (5′-GATTAATTAACTCACTGCAGTCTTCTATAACT-3′) (SEQ ID NO:93) respectively, introduced AscI and Pacd restriction sites are underlined).
  • the catalytic domain of rat GnTIl was than cloned downstream of the PMA1 promoter as a AscI-Pacl fragment in pBP 124.
  • the gene fragment encoding the S. cerevisiae Mnn2 localization signal was cloned from pJN281 as a NotI-AscI fragment to generate an in-frame fusion with the catalytic domain of GnTII, to generate plasmid pTC53.
  • N-acetylglucosamine to the GlcNAc 2 Man 3 GlcNAc 2 structure by N-acetylglucosaminyltransferases III yields a so-called bisected N-glycan (see FIG. 15 ).
  • This structure has been implicated in greater antibody-dependent cellular cytotoxicity (ADCC) (Umana et al. (1999) Nat. Biotechnol. 17(2):176-80).
  • a host cell such as a yeast strain capable of producing glycoproteins with bisected N-glycans is engineered according to the invention, by introducing into the host cell a GnTIII activity.
  • the host cell is transformed with a nucleic acid that encodes GnTIII (e.g., a mammalian such as the murine GnTIII shown in FIG. 24 ) or a domain thereof having enzymatic activity, optionally fused to a heterologous cell signal targeting peptide (e.g., using the libraries and associated methods of the invention.)
  • IgGs consist of two heavy-chains (V H , C H 1, C H 2 and C H 3 in FIG. 22 ), interconnected in the hinge region through three disulfide bridges, and two light chains (V L , C L in FIG. 22 ).
  • the light chains domains V L and C L
  • the Fc region of IgGs consists of the C H 3, the C H 2 and the hinge region and is responsible for the exertion of so-called effector functions (see below).
  • Antibodies of the IgG subclass exert two major effector-functions: the activation of the complement system and induction of phagocytosis.
  • the complement system consists of a complex group of serum proteins involved in controlling inflammatory events, in the activation of phagocytes and in the lytical destruction of cell membranes. Complement activation starts with binding of the C1 complex to the Fc portion of two IgGs in close proximity.
  • C1 consists of one molecule, C1q, and two molecules, C1r and C1s.
  • Phagocytosis is initiated through an interaction between the IgG's Fc fragment and Fc-gamma-receptors (Fc ⁇ RI, II and III in FIG. 22 ).
  • Fc receptors are primarily expressed on the surface of effector cells of the immune system, in particular macrophages, monocytes, myeloid cells and dendritic cells.
  • the C H 2 portion harbors a conserved N-glycosylation site at asparagine 297 (Asn297).
  • the Asn297 N-glycans are highly heterogeneous and are known to affect Fc receptor binding and complement activation. Only a minority (i.e., about 15-20%) of IgGs bears a disialylated, and 3-10% have a monosialylated N-glycan (reviewed in Jefferis (2001) Biopharm. 14:19-26).
  • N-glycan structure shown to be necessary for fully functional antibodies capable of complement activation and Fc receptor binding is a pentasacharide with terminal N-acetylglucosamine residues (GlcNAc 2 Man 3 ) (reviewed in Jefferis, R., Glycosylation of human IgG Antibodies. BioPharm, 2001).
  • Antibodies with less than a GlcNAc 2 Man 3 N-glycan or no N-glycosylation at Asn297 might still be able to bind an antigen but most likely will not activate the crucial downstream events such as phagocytosis and complement activation.
  • antibodies with fungal-type N-glycans attached to Asn297 will in all likelihood solicit an immune-response in a mammalian organism which will render that antibody useless as a therapeutic glycoprotein.
  • the DNA fragment encoding part of the mouse GnTIII protein lacking the TM domain is PCR amplified from murine (or other mammalian) genomic DNA using forward (5′-TCCTGGCGCGCCTTCCCGAGAGAACTGGCCTCCCTC-3′) (SEQ ID NO:94) and reversed (5′-AATTAATTAACCCTAGCCCTCCGCTGTATCCAACTTG-3′) (SEQ ID NO:95) primers.
  • Those primers include AscI and Pacl restriction sites that may be used for cloning into the vector suitable for the fusion with leader library.
  • the nucleic acid (SEQ ID NO:45) and amino acid (SEQ ID NO:46) sequences of murine GnTIII are shown in FIG. 24 .
  • Sources of antibodies and encoding genes can be, among others, in vitro immunized human B cells (see, e.g., Borreback et al. (1988) Proc. Natl. Acad. Sci. USA 85:3995-3999), peripheral blood lymphocytes or single human B cells (see, e.g., Lagerkvist et al. (1995) Biotechniques 18:862-869; and Terness et al. (1997) Hum. Immunol. 56:17-27) and transgenic mice containing human immunoglobulin loci, allowing the creation of hybridoma cell-lines.
  • antibody-encoding nucleic acid sequences can be cloned.
  • Sources for the genetic information encoding immunoglobulins of interest are typically total RNA preparations from cells of interest, such as blood lymphocytes or hybridoma cell lines.
  • variable regions can be cloned via reverse transcription initiated from a sequence-specific primer hybridizing to the IgG C H 1 domain site and a second primer encoding amino acids 111-118 of the murine kappaconstant region.
  • the V H and V K encodinWDNAs can then be amplified as previously published (see, e.g., Graziano et al. (1995) J.
  • FIG. 23 shows a schematic overview of such an expression construct, designed in a very modular way, allowing easy exchange of promoters, transcriptional terminators, integration targeting domains and even selection markers.
  • V L as well as V H domains of choice can be easily cloned in-frame with C L and the C H regions, respectively.
  • Initial integration is targeted to the P. pastoris AOX locus (or homologous locus in another fungal cell) and the methanol-inducible AOX promoter will drive expression.
  • any other desired constitutive or inducible promoter cassette may be used.
  • the 5′ AOX and 3′ AOX regions as well as transcriptional terminator (TT) fragments can be easily replaced with different TT, promoter and integration targeting domains to optimize expression.
  • the alpha-factor secretion signal with the standard KEX protease site is employed to facilitate secretion of heavy and light chains.
  • the properties of the expression vector may be further refined using standard techniques.
  • An Ig expression vector such as the one described above is introduced into a host cell of the invention that expresses GnTIII, preferably in the Golgi apparatus of the host cell.
  • the Ig molecules expressed in such a host cell comprise N-glycans having bisecting GlcNAcs.
  • the previously reported P. pastoris strain BK64 (Choi et al. (2003) Proc Natl Acad Sci USA. 100(9):5022-7), a triple auxotroph (ADE, ARG, HIS) possessing the OCH1 knock-out and expressing the kringle 3 domain (K3) of human plasminogen, was used as the host strain.
  • BK64 was transformed with the plasmid pPB 103 linearized with the restriction enzyme EcoNI to introduce the K. lactis UDP-N-acetylglucosamine transporter into the host cell, thus creating the strain PBP-1.
  • the mouse Mnsl was introduced into this strain by transformation with the plasmid pFB8 linearized with the restriction enzyme EcoNI, generating strain PBP-2.
  • K3 glycan analysis from proteins isolated from strain PBP-2 demonstrated that the primary glycoform present was Man 5 GlcNAc 2 .
  • PBP-2 was subsequently transformed with the human GnTI plasmid pNA15 linearized with the restriction enzyme AatII, generating the strain PBP-3.
  • Analysis of the K3 glycoforms produced in strain PBP-3 demonstrated that the hybrid glycan GlcNAcMan 5 GlcNAc 2 was the predominant structure.
  • this strain was grown in YPD prior to selection on minimal media containing 5-Fluoroorotic (5-FOA, BioVectra) and uracil (Boeke et al. (1984) Mol. Gen. Genet. 197:345-346).
  • the recovered Ura-minus strain producing GlcNAcMan 5 GlcNAc 2 glycoforms was designated YSH-1 ( FIG. 36 ).
  • the N-glycan profile from strain YSH-1 is shown in FIG. 25 (top) and displays a predominant peak at 1465 m/z corresponding to the mass of GlcNAcMan 5 GlcNAc 2 [d].
  • YSH-1 (Example 13) was transformed with the D. melanogaster mannosidase II ⁇ 74/ S. cerevisiae MNN2(s) plasmid (pKD53) linearized with the restriction enzyme ApaI, generating strain YSH-37 ( FIG. 36 ).
  • Analysis of the K3 glycan structures produced in strain YSH-37 ( FIG. 25 (bottom)) demonstrated that the predominant glycoform at 1140 m/z corresponds to the mass of GlcNAcMan 3 GlcNAc 2 [b] and other glycoforms GlcNAcMan 4 GlcNAc 2 [c] at 1303 m/z and GlcNAcMan 5 GlcNAc 2 [d] at 1465 m/z.
  • Strain YSH-37 (Example 14) was transformed with a plasmid encoding a rat GnTII/MNN2 (s) leader, pTC53, linearized with the restriction enzyme EcoRI.
  • the resulting strain, YSH-44 ( FIG. 36 ), produced a K3 N-glycan having a single glycoform at 1356 m/z, corresponding to the mass of GlcNAc 2 Man 3 GlcNAc 2 [x], by positive mode MALDI-TOF mass spectrometry ( FIG. 29 ).
  • the plasmid pBLURA-SX (from Jim Cregg) was digested with BamHI and BglII to release the AOXexpression cassette.
  • the plasmid pJN338 was cut with NotI and PacI and the two oligonucleotides 5′-GGCCGCCTGCAGATTTAAATGAATTCGGCGCGCCTTAAT-3′ (SEQ ID NO:96) and 5′-TAAGGCGCGCC GAATTCATTTAAATCTGCAGGGC-3′ (SEQ ID NO:97) that had been annealed in vitro, were ligated into the open sites, to create pJN348.
  • the PpURA3 containing GAPDH expression vector pJN348 was linearized with XhoI and blunted with T4 DNA polymerase and calf intestinal phosphatase (CIP) treated.
  • the HYG resistance marker was digested from pAG32 with BglII and SacI and blunted, then ligated into pJN348 to create pRCD259 which can be used as a HYG expression vector that integrates at the PpURA3 locus.
  • the PCR products were then cloned into pJN 348 vector as AscI-Pacl fragments and sequenced.
  • the resulting vectors pVA (GnTIII ⁇ 32), pVB (GnTIII ⁇ 86), and pVC (GnTIII ⁇ 212) were digested with NotI-AscI enzymes and used for the ligation with yeast leader library (leaders 20-67).
  • These targeting peptides are fused to the catalytic domains selected from the mouse GnTIII with 32, 86, 212 amino acid N-terminal deletions.
  • the MNN2 targeting peptide from S. cerevisiae (long, medium and short) and GNT1 from K.
  • lactis (short, and medium) (see Example 11) are shown in Table 11.
  • Table 11 A representative combinatorial library of targeting peptide sequences/catalytic domains exhibiting UDP-N- Acetylglucosaminyltransferase III (GnTIII) activity in P. pastoris YSH-1 Targeting peptide S. cerevisiae S. cerevisiae S. cerevisiae K.
  • the P. pastoris strain producing GlcNAcMan 5 GlcNAc 2 (PBP-3) (see Example 8) was counterselected on 5-FOA, thereby selecting for loss of the URA3+ marker and a ura3 ⁇ phenotype.
  • This strain, designated YSH-1 ( FIG. 36 ), was transformed with the library of N-acetylglucosaminyltransferase III (GnTIII) catalytic domains (vectors pVA, pVB, and pVC) and leaders. Transformants were grown at 30° C.
  • K3 Kringle 3 from human plasminogen
  • K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot.
  • the robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3).
  • the N-glycans were released by PNGase digestion (Example 3).
  • the N-glycans were analyzed with a MALDI-TOF MS (Example 3).
  • the GnTIII activities are shown in Table 11.
  • (+)s indicates the relative levels of bisected N-glycan production of % neutral glycans.
  • Targeting peptide sequences were selected from selected from the group consisting of: Saccharomyces GLS1, Saccharomyces MNS1, Saccharomyces SEC12, Pichia SEC, Pichia OCH 1, Saccharomyces MNN9, Saccharomyces VAN 1, Saccharomyces ANP1, Saccharomyces HOC1, Saccharomyces MNN10, Saccharomyces MNN11, Saccharomyces MNT1, Pichia D2, Pichia D9, Pichia J3, Saccharomyces KTR1 , Saccharomyces KTR2, Kluyveromyces GnTI, Saccharomyces MNN2, Saccharomyces MNN5, Saccharomyces YUR1, Saccharomyces MNN1, and Saccharomyces MNN6.
  • GnTIII constructs from vectors pVA53, pVB53, pVA54, and pVB54 were transferred as NotI-PacI fragments into pRCD259 to generate vectors pPB135, pPB137, pPB136, and pPB138.
  • the vectors contain HYG resistance marker and P. pastoris URA3 gene as targeting sequence for genomic integration.
  • Plasmids are linearized with SalI, transformed into strain YSH-44 by electroporation, selected on medium containing hygromycin and the resulting strains are screened by analysis of the released glycans from purified K3. Transformants were grown at 24° C.
  • K3 Kringle 3 from human plasminogen
  • K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well forrnat on a Beckman BioMek 2000 laboratory robot (Example 3).
  • the robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3).
  • the N-glycans were released by PNGase digestion.
  • the N-glycans were analyzed with a MALDI-TOF MS (Example 3).
  • the pPB135 transformants exhibiting the bisecting GlcNAc (e.g. GlcNAc 2 Man 5 GlcNAc 2 ) were designated YSH-57 ( FIG. 36 ).
  • Table 11 depicts the activity of the mouse GnTIII.
  • the P. pastoris PBP6-5 (Example 11) was transformed with the plasmid pPB135 (Table 11) encoding a mouse GnTIII catalytic domain ( ⁇ 32) ligated in frame to a targeting peptide derived from S. cerevisiae MNN2. Transformants were grown at 30° C. in BMGY to an OD600 of about 10, harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot.
  • the robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3).
  • the N-glycans were released by PNGase digestion (Example 3).
  • the N-glycans were analyzed with a MALDI-TOF MS (Example 3).
  • Transformants exhibiting the bisecting GlcNAc e.g. GlcNAc 2 Man 3 GlcNAc 2
  • PBP-38 FIG. 36
  • Table 11 depicts the activity of the mouse GnTIII.
  • YSH-57 cell culture supernatants were tested for GnTIII activity.
  • P. pastoris YSH-57 cells were grown at 24° C. in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for GnTIII assays and the remainder was used for the recovery of secreted soluble K3.
  • K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot.
  • the robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3).
  • the N-glycans were released by PNGase digestion (Example 3).
  • the earlier removed aliquot of the supernatant was further tested for the presence of secreted GnTIII activity.
  • GlcNAcMan 5 GlcNAc 2 purified from K3 expressed in PBP-3 strain was added to: BMMY (A) 1 mM UDP-GlcNAc (Sigma Chemical Co., St.
  • P. pastoris YSH-57 cells culture supernatants were tested for GnTIII activity.
  • P. pastoris YSH-57 cells were grown at 24° C. in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for GnTIII assays and the remainder was used for the recovery of secreted soluble K3.
  • K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot.
  • the robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3).
  • the N-glycans were released by PNGase digestion (Example 3).
  • the earlier removed aliquot of the supernatant was further tested for the presence of secreted GnTIII activity.
  • GlcNAc 2 Man 3 GlcNAc 2 purified from K3 expressed in YSH-44 strain was added to: BMMY (A) 1 mM UDP-GlcNAc (Sigma Chemical Co., St.

Abstract

The present invention relates to eukaryotic host cells having modified oligosaccharides which may be modified further by heterologous expression of a set of glycosyltransferases, sugar transporters and mannosidases to become host-strains for the production of mammalian, e.g., human therapeutic glycoproteins. The process provides an engineered host cell which can be used to express and target any desirable gene(s) involved in glycosylation. Host cells with modified lipid-linked oligosaccharides are created or selected. N-glycans made in the engineered host cells exhibit GnTIII activity, which produce bisected N-glycan structures and may be modified further by heterologous expression of one or more enzymes, e.g., glycosyltransferases, sugar transporters and mannosidases, to yield human-like glycoproteins. For the production of therapeutic proteins, this method may be adapted to engineer cell lines in which any desired glycosylation structure may be obtained.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of U.S. application Ser. No. 10/371,877, filed on Feb. 20, 2003, which is a continuation-in-part of U.S. application Ser. No. 09/892,591, filed Jun. 27, 2001, which claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 60/214,358, filed Jun. 28, 2000, U.S. Provisional Application No. 60/215,638, filed Jun. 30, 2000, and U.S. Provisional Application No. 60/279,997, filed Mar. 30, 2001, each of which is incorporated herein by reference in its entirety. This application is also a continuation-in-part of PCT/US02/41510, filed on Dec. 24, 2002, which claims the benefit of U.S. Provisional Application No. 60/344,169, filed on Dec. 27, 2001, each of which is incorporated herein by reference in its entirety.
  • FIELD OF THE INVENTION
  • The present invention is directed to methods and compositions by which non-human eukaryotic host cells, such as fungi or other eukaryotic cells, can be genetically modified to produce glycosylated proteins (glycoproteins) having patterns of glycosylation similar to those of glycoproteins produced by animal cells, especially human cells, which are useful as human or animal therapeutic agents.
  • BACKGROUND OF THE INVENTION
  • Glycosylation Pathways in Humans and Lower Eukaryotes
  • After DNA is transcribed and translated into a protein, further post-translational processing involves the attachment of sugar residues, a process known as glycosylation. Different organisms produce different glycosylation enzymes (glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available, so that the glycosylation patterns as well as composition of the individual oligosaccharides, even of the same protein, will be different depending on the host system in which the particular protein is being expressed. Bacteria typically do not glycosylate proteins, and if so only in a very unspecific manner (Moens and Vanderleyden (1997) Arch Microbiol. 168(3):169-175). Lower eukaryotes such as filamentous fungi and yeast add primarily mannose and mannosylphosphate sugars. The resulting glycan is known as a “high-mannose” type glycan or a mannan. Plant cells and insect cells (such as Sf9 cells) glycosylate proteins in yet another way. By contrast, in higher eukaryotes such as humans, the nascent oligosaccharide side chain may be trimmed to remove several mannose residues and elongated with additional sugar residues that typically are not found in the N-glycans of lower eukaryotes. See, e.g., Bretthauer, et al. (1999) Biotechnology and Applied Biochemistry 30:193-200; Martinet, et al. (1998) Biotechnology Letters 20:1171-1177; Weikert, et al. (1999) Nature Biotechnology, 17:1116-1121; M. Malissard, et al. (2000) Biochemical and Biophysical Research Communications 267:169-173; Jarvis, etal., (1998) Current Opinion in Biotechnology 9:528-533; and Takeuchi (1997) Trends in Glycoscience and Glycotechnology 9:S29-S35.
  • Synthesis of a mammalian-type oligosaccharide structure begins with a set of sequential reactions in the course of which sugar residues are added and removed while the protein moves along the secretory pathway in the host organism. The enzymes which reside along the glycosylation pathway of the host organism or cell determine the resulting glycosylation patterns of secreted proteins. Thus, the resulting glycosylation pattern of proteins expressed in lower eukaryotic host cells differs substantially from the glycosylation pattern of proteins expressed in higher eukaryotes such as humans and other mammals (Bretthauer, 1999). The structure of a typical fungal N-glycan is shown in FIG. 1A.
  • The early steps of human glycosylation can be divided into at least two different phases: (i) lipid-linked Glc3Man9GlcNAc2 oligosaccharides are assembled by a sequential set of reactions at the membrane of the endoplasmic reticulum (ER) (FIG. 13) and (ii) the transfer of this oligosaccharide from the lipid anchor dolichyl pyrophosphate onto de novo synthesized protein. The site of the specific transfer is defined by an asparagine (Asn) residue in the sequence Asn-Xaa-Ser/Thr (SEQ ID NOs:1 and 2) where Xaa can be any amino acid except proline (Gavel and von Heijne (1990) Protein Eng. 3:433-42). Further processing by glucosidases and mannosidases occurs in the ER before the nascent glycoprotein is transferred to the early Golgi apparatus, where additional mannose residues are removed by Golgi specific alpha (α)-1,2-mannosidases. Processing continues as the protein proceeds through the Golgi. In the medial Golgi, a number of modifying enzymes, including N-acetylglucosaminyl transferases (GnTI, GnTII, GnTIII, GnTIV and GnTV), mannosidase II and fucosyltransferases, add and remove specific sugar residues. Finally, in the trans-Golgi, galactosyltranferases (GalT) and sialyltransferases (ST) produce a glycoprotein structure that is released from the Golgi. It is this structure, characterized by bi-, tri- and tetra-antennary structures, containing galactose, fucose, N-acetylglucosamine and a high degree of terminal sialic acid, that gives glycoproteins their human characteristics. The structure of a typical human N-glycan is shown in FIG. 1B. See also FIGS. 14 and 15 for steps involved in mammalian-type N-glycan processing.
  • In nearly all eukaryotes, glycoproteins are derived from a common lipid-linked oligosaccharide precursor Glc3Man9GlcNAc2-dolichol-pyrophosphate. Within the endoplasmic reticulum, synthesis and processing of dolichol pyrophosphate bound oligosaccharides are identical between all known eukaryotes. However, further processing of the core oligosaccharide by fungal cells, e.g., yeast, once it has been transferred to a peptide leaving the ER and entering the Golgi, differs significantly from humans as it moves along the secretory pathway and involves the addition of several mannose sugars.
  • In yeast, these steps are catalyzed by Golgi residing mannosyltransferases, like Och1p, Mnt1p and Mnn1p, which sequentially add mannose sugars to the core oligosaccharide. The resulting structure is undesirable for the production of human-like proteins and it is thus desirable to reduce or eliminate mannosyltransferase activity. Mutants of S. cerevisiae, deficient in mannosyltransferase activity (for example och1 or mnn9 mutants) have been shown to be non-lethal and display reduced mannose content in the oligosaccharide of yeast glycoproteins. Other oligosaccharide processing enzymes, such as mannosylphosphate transferase, may also have to be eliminated depending on the host's particular endogenous glycosylation pattern.
  • Sugar Nucleotide Precursors
  • The N-glycans of animal glycoproteins typically include galactose, fucose, and terminal sialic acid. These sugars are not found on glycoproteins produced in yeast and filamentous fungi. In humans, the full range of nucleotide sugar precursors (e.g., UDP-N-acetylglucosamine, UDP-N-acetylgalactosamine, CMP-N-acetylneuraminic acid, UDP-galactose, GDP-fucose, etc.) are synthesized in the cytosol and transported into the Golgi, where they are attached to the core oligosaccharide by glycosyltransferases. (Sommers and Hirschberg (1981) J. Cell Biol. 91(2):A406-A406; Sommers and Hirschberg (1982) J. Biol. Chem. 257(18):811-817; Perez and Hirschberg (1987) Methods in Enzymology 138:709-715).
  • Glycosyl transfer reactions typically yield a side product which is a nucleoside diphosphate or monophosphate. While monophosphates can be directly exported in exchange for nucleoside triphosphate sugars by an antiport mechanism, diphosphonucleosides (e.g., GDP) have to be cleaved by phosphatases (e.g. GDPase) to yield nucleoside monophosphates and inorganic phosphate prior to being exported. This reaction is important for efficient glycosylation; for example, GDPase from Saccharomyces cerevisiae (S. cerevisiae) has been found to be necessary for mannosylation. However that GDPase has 90% reduced activity toward UDP (Berninsone et al. (1994) J. Biol. Chem. 269(1):207-21 1). Lower eukaryotes typically lack UDP-specific diphosphatase activity in the Golgi since they do not utilize UDP-sugar precursors for Golgi-based glycoprotein synthesis. Schizosaccharomyces pombe, a yeast found to add galactose residues to cell wall polysaccharides (from UDP-galactose) has been found to have specific UDPase activity, indicating the potential requirement for such an enzyme (Berninsone et al. (1994) J. Biol. Chem. 269(l):207-211). UDP is known to be a potent inhibitor of glycosyltransferases and the removal of this glycosylation side product may be important to prevent glycosyl-transferase inhibition in the lumen of the Golgi (Khatara et al. (1974) Eur. J. Biochem. 44:537-560). See Berninsone et al. (1995) J. Biol. Chem. 270(24):14564-14567; Beaudet et al. (1998) Abc Transporters: Biochemical, Cellular, and Molecular Aspects 292: 397-413.
  • Sequential Processing of N-glycans by Compartmentalized Enzyme Activities
  • Sugar transferases and glycosidases (e.g., mannosidases) line the inner (luminal) surface of the ER and Golgi apparatus and thereby provide a “catalytic” surface that allows for the sequential processing of glycoproteins as they proceed through the ER and Golgi network. The multiple compartments of the cis, medial, and trans Golgi and the trans-Golgi Network (TGN), provide the different localities in which the ordered sequence of glycosylation reactions can take place. As a glycoprotein proceeds from synthesis in the ER to full maturation in the late Golgi or TGN, it is sequentially exposed to different glycosidases, mannosidases and glycosyltransferases such that a specific carbohydrate structure may be synthesized. Much work has been dedicated to revealing the exact mechanism by which these enzymes are retained and anchored to their respective organelle. The evolving picture is complex but evidence suggests that stem region, membrane spanning region and cytoplasmic tail, individually or in concert, direct enzymes to the membrane of individual organelles and thereby localize the associated catalytic domain to that locus (see, e.g., Gleeson (1998) Histochem. Cell Biol. 109:517-532).
  • In some cases, these specific interactions were found to function across species. For example, the membrane spanning domain of α2,6-ST from rats, an enzyme known to localize in the trans-Golgi of the animal, was shown to also localize a reporter gene (invertase) in the yeast Golgi (Schwientek et al. (1995) J. Biol. Chem. 270(10):5483-9). However, the very same membrane spanning domain as part of a full-length α2,6-ST was retained in the ER and not further transported to the Golgi of yeast (Krezdorn et al. (1994) Eur. J. Biochem. 220(3):809-17). A full length GalT from humans was not even synthesized in yeast, despite demonstrably high transcription levels. In contrast, the transmembrane region of the same human GalT fused to an invertase reporter was able to direct localization to the yeast Golgi, albeit it at low production levels. Schwientek and co-workers have shown that fusing 28 amino acids of a yeast mannosyltransferase (MNT1), a region containing a cytoplasmic tail, a transmembrane region and eight amino acids of the stem region, to the catalytic domain of human GalT are sufficient for Golgi localization of an active GalT. Other galactosyltransferases appear to rely on interactions with enzymes resident in particular organelles because, after removal of their transmembrane region, they are still able to localize properly.
  • Improper localization of a glycosylation enzyme may prevent proper functioning of the enzyme in the pathway. For example, Aspergillus nidulans, which has numerous α-1,2-mannosidases (Eades and Hintz (2000) Gene 255(l):25-34), does not add GlcNAc to Man5GlcNAc2 when transformed with the rabbit GnTI gene, despite a high overall level of GnTI activity (Kalsner et al. (1995) Glycoconj. J. 12(3):360-370). GnTI, although actively expressed, may be incorrectly localized such that the enzyme is not in contact with both of its substrates: UDP-GlcNAc and a productive Man5GlcNAc2 substrate (not all Man5GlcNAc2 structures are productive; see below). Alternatively, the host organism may not provide an adequate level of UDP-GlcNAc in the Golgi or the enzyme may be properly localized but nevertheless inactive in its new environment. In addition, Man5GlcNAc2 structures present in the host cell may differ in structure from Man5GlcNAc2 found in mammals. Maras and coworkers found that about one third of the N-glycans from cellobiohydrolase I (CBHI) obtained from T. reesei can be trimmed to Man5GlcNAc2 by A. saitoi 1,2 mannosidase in vitro. Fewer than 1% of those N-glycans, however, could serve as a productive substrate for GnTI. Maras et al. (1997) Eur. J. Biochem. 249:701-707. The mere presence of Man5GlcNAc2, therefore, does not assure that further in vivo processing of Man5GlcNAc2 can be achieved. It is formation of a productive, GnTI-reactive Man5GlcNAc2 structure that is required. Although Man5GlcNAc2 could be produced in the cell (about 27 mol %), only a small fraction could be converted to Man5GlcNAc2 (less than about 5%, see Chiba et al. WO 01/14522).
  • To date, there is no reliable way of predicting whether a particular heterologously expressed glycosyltransferase or mannosidase in a lower eukaryote will be (1), sufficiently translated (2), catalytically active or (3) located to the proper organelle within the secretory pathway. Because all three of these are necessary to affect glycosylation patterns in lower eukaryotes, a systematic scheme to achieve the desired catalytic function and proper retention of enzymes in the absence of predictive tools, which are currently not available, would be desirable.
  • Production of Therapeutic Glycoproteins
  • A significant number of proteins isolated from humans or animals are post-translationally modified, with glycosylation being one of the most significant modifications. An estimated 70% of all therapeutic proteins are glycosylated and thus currently rely on a production system (i.e., host cell) that is able to glycosylate in a manner similar to humans. Several studies have shown that glycosylation plays an important role in determining the (1) immunogenicity, (2) pharmacokinetic properties, (3) trafficking, and (4) efficacy of therapeutic proteins. It is thus not surprising that substantial efforts by the pharmaceutical industry have been directed at developing processes to obtain glycoproteins that are as “humanoid” or “human-like” as possible. To date, most glycoproteins are made in a mammalian host system. This may involve the genetic engineering of such mammalian cells to enhance the degree of sialylation (i.e., terminal addition of sialic acid) of proteins expressed by the cells, which is known to improve pharmacokinetic properties of such proteins. Alternatively, one may improve the degree of sialylation by in vitro addition of such sugars using known glycosyltransferases and their respective nucleotide sugars (e.g., 2,3-sialyltransferase and CMP-sialic acid).
  • While most higher eukaryotes carry out glycosylation reactions that are similar to those found in humans, recombinant human proteins expressed in the above mentioned host systems invariably differ from their “natural” human counterpart (Raju et al. (2000) Glycobiology 10(5): 477-486). Extensive development work has thus been directed at finding ways to improve the “human character” of proteins made in these expression systems. This includes the optimization of fermentation conditions and the genetic modification of protein expression hosts by introducing genes encoding enzymes involved in the formation of human-like glycoforms. Goochee et al. (1999) Biotechnology 9(12):1347-55; Andersen and Goochee (1994) Curr Opin Biotechnol. 5(5):546-49; Werner et al. (1998) Arzneimittelforschung. 48(8):870-80; Weikert et al. (1999) Nat Biotechnol. 17(11):1116-21; Yang and Butler (2000) Biotech. Bioeng. 68:370-80. Inherent problems associated with all mammalian expression systems have not been solved.
  • Glycoprotein Production Using Eukaryotic Microorganisms
  • Although the core oligosaccharide structure transferred to a protein in the endoplasmic reticulum is basically identical in mammals and lower eukaryotes, substantial differences have been found in the subsequent processing reactions which occur in the Golgi apparatus of fungi and mammals. In fact, even amongst different lower eukaryotes there exist a great variety of glycosylation structures. This has historically prevented the use of lower eukaryotes as hosts for the production of recombinant human glycoproteins despite otherwise notable advantages over mammalian expression systems.
  • Therapeutic glycoproteins produced in a microorganism host such as yeast utilizing the endogenous host glycosylation pathway differ structurally from those produced in mammalian cells and typically show greatly reduced therapeutic efficacy. Such glycoproteins are typically immunogenic in humans and show a reduced half-life (and thus bioactivity) in vivo after administration (Takeuchi (1997) Trends in Glycoscience and Glycotechnology 9:S29-S35). Specific receptors in humans and animals (i.e., macrophage mannose receptors) can recognize terminal mannose residues and promote the rapid clearance of the foreign glycoprotein from the bloodstream. Additional adverse effects may include changes in protein folding, solubility, susceptibility to proteases, trafficking, transport, compartmentalization, secretion, recognition by other proteins or factors, antigenicity, or allergenicity.
  • Yeast and filamentous fungi have both been successfully used for the production of recombinant proteins, both intracellular and secreted (Cereghino and Cregg (2000) FEMS Microbiology Reviews 24(1):45-66; Harkki et al. (1989) Bio-Technology 7(6):596; Berka et al. (1992) Abstr. Papers Amer. Chem. Soc. 203:121-BIOT; Svetina et al. (2000) J. Biotechnol. 76(2-3):245-251). Various yeasts, such as K. lactis, Pichia pastoris, Pichia methanolica, and Hansenula polymorpha, have played particularly important roles as eukaryotic expression systems because they are able to grow to high cell densities and secrete large quantities of recombinant protein. Likewise, filamentous fungi, such as Aspergillus niger, Fusarium sp, Neurospora crassa and others, have been used to efficiently produce glycoproteins at the industrial scale. However, as noted above, glycoproteins expressed in any of these eukaryotic microorganisms differ substantially in N-glycan structure from those in animals. This has prevented the use of yeast or filamentous fungi as hosts for the production of many therapeutic glycoproteins.
  • Although glycosylation in yeast and fungi is very different than in humans, some common elements are shared. The first step, the transfer of the core oligosaccharide structure to the nascent protein, is highly conserved in all eukaryotes including yeast, fungi, plants and humans (compare FIGS. 1A and 1B). Subsequent processing of the core oligosaccharide, however, differs significantly in yeast and involves the addition of several mannose sugars. This step is catalyzed by mannosyltransferases residing in the Golgi (e.g., OCH1, MNT1, MNN1, etc.), which sequentially add mannose sugars to the core oligosaccharide. The resulting structure is undesirable for the production of humanoid proteins and it is thus desirable to reduce or eliminate mannosyltransferase activity. Mutants of S. cerevisiae deficient in mannosyltransferase activity (e.g., och1 or mnn9 mutants) have shown to be non-lethal and display a reduced mannose content in the oligosaccharide of yeast glycoproteins. Other oligosaccharide processing enzymes, such as mannosylphosphate transferase, may also have to be eliminated depending on the host's particular endogenous glycosylation pattern. After reducing undesired endogenous glycosylation reactions, the formation of complex N-glycans has to be engineered into the host system. This requires the stable expression of several enzymes and sugar-nucleotide transporters. Moreover, one has to localize these enzymes so that a sequential processing of the maturing glycosylation structure is ensured.
  • Several efforts have been made to modify the glycosylation pathways of eukaryotic microorganisms to provide glycoproteins more suitable for use as mammalian therapeutic agents. For example, several glycosyltransferases have been separately cloned and expressed in S. cerevisiae (GalT, GnTI), Aspergillus nidulans (GnTI) and other fungi (Yoshida et al. (1999) Glycobiology 9(1):53-8, Kalsner et al. (1995) Glycoconj. J 12(3):360-370). However, N-glycans resembling those made in human cells were not obtained.
  • Yeasts produce a variety of mannosyltransferases (e.g., 1,3-mannosyltransferases such as MNN1 in S. cerevisiae; Graham and Emr (1991) J. Cell. Biol. 114(2):207-218), 1,2-mannosyltransferases (e.g., KTR/KRE family from S. cerevisiae), 1,6-mannosyltransferases (e.g., OCH1 from S. cerevisiae), mannosylphosphate transferases and their regulators (e.g., MNN4 and MNN6 from S. cerevisiae) and additional enzymes that are involved in endogenous glycosylation reactions. Many of these genes have been deleted individually giving rise to viable organisms having altered glycosylation profiles. Examples are shown in Table 1.
    TABLE 1
    Examples of yeast strains having altered mannosylation
    Strain N-glycan (wild type) Mutation N-glycan (mutant) Reference
    S. pombe Man>9GlcNAc2 OCH1 Man8GlcNAc2 Yoko-o et al.
    (2001) FEBS Lett.
    489(1): 75-80
    S. cerevisiae Man>9GlcNAc2 OCH1/MNN1 Man8GlcNAc2 Nakanishi-Shindo
    et al. (1993) J.
    Biol. Chem.
    268(35): 26338-26345
    S. cerevisiae Man>9GlcNAc2 OCH1/MNN1/MNN4 Man8GlcNAc2 Chiba et al. (1998)
    J. Biol. Chem.
    273, 26298-26304
    P. pastoris Hyperglycosylated OCH1 (complete Not Welfide, Japanese
    deletion) hyperglycosylated Application
    Publication No. 8-
    336387
    P. pastoris Man>8GlcNAc2 OCH1 (disruption) Man>8GlcNAc2 Contreras et al.
    WO 02/00856 A2
  • Japanese Patent Application Publication No. 8-336387 discloses the deletion of an OCH1 homolog in Pichia pastoris. In S. cerevisiae, OCH1 encodes a 1,6-mannosyltransferase, which adds a mannose to the glycan structure Man8GlcNAc2 to yield Man9GlcNAc2. The Man9GlcNAc2 structure, which contains three 1,6 mannose residues, is then a substrate for further 1,2-, 1,6-, and 1,3- mannosyltransferases in vivo, leading to the hypermannosylated glycoproteins that are characteristic for S. cerevisiae and which typically may have 30-40 mannose residues per N-glycan. Because the Och1p initiates the transfer of 1,6 mannose to the Man8GlcNAc2 core, it is often referred to as the “initiating 1,6 mannosyltransferase” to distinguish it from other 1,6 mannosyltransferases acting later in the Golgi. In an och1 mnn1 mnn4 mutant strain of S. cerevisiae, proteins glycosylated with Man8GlcNAc2 accumulate and hypermannosylation does not occur. However, Man8GlcNAc2 is not a substrate for mammalian glycosyltransferases, such as human UDP-GlcNAc transferase I, and accordingly, the use of that mutant strain, in itself, is not useful for producing mammalian-like proteins, i.e., those with complex or hybrid glycosylation patterns.
  • One can trim Man8GlcNAc2 structures to a Man5GlcNAc2 isomer in S. cerevisiae (although high efficiency trimming greater than 50% in vivo has yet to be demonstrated) by engineering a fungal mannosidase from A. saitoi into the endoplasmic reticulum (ER). The shortcomings of this approach are two-fold: (1) it is not clear whether the Man5GlcNAc2 structures formed are in fact formed in vivo (rather than having been secreted and further modified by mannosidases outside the cell); and (2) it is not clear whether any Man5GlcNAc2 structures formed, if in fact formed in vivo, are the correct isoform to be a productive substrate for subsequent N-glycan modification by GlcNAc transferase I (Maras et al. (1997) Eur. J Biochem. 249:701-707).
  • With the objective of providing a more human-like glycoprotein derived from a fungal host, U.S. Pat. No. 5,834,251 discloses a method for producing a hybrid glycoprotein derived from Trichoderma reseei. A hybrid N-glycan has only mannose residues on the Manα1-6 arm of the core mannose structure and one or two complex antennae on the Manα1-3 arm. While this structure has utility, the method has the disadvantage that numerous enzymatic steps must be performed in vitro, which is costly and time-consuming. Isolated enzymes are expensive to prepare and need costly substrates (e.g., UDP-GlcNAc). The method also does not allow for the production of complex glycans on a desired protein.
  • Intracellular Mannosidase Activity Involved in N-glycan Trimming
  • Alpha-1,2-mannosidase activity is required for the trimming of Man8GlcNAc2 to form Man5GlcNAc2, which is a major intermediate for complex N-glycan formation in mammals. Previous work has shown that truncated murine, fungal and human α-1,2-mannosidase can be expressed in the methylotropic yeast P. pastoris and display Man8GlcNAc2 to Man5GlcNAc2 trimming activity (Lal et al. (1998) Glycobiology 8(10):981-95; Tremblay et al. (1998) Glycobiology 8(6):585-95, Callewaert et al. (2001) FEBS Lett. 503(2-3):173-8). However, to date, no reports exist that show the high level in vivo trimming of Man8GlcNAc2 to Man5GlcNAc2 on a secreted glycoprotein from P. pastoris.
  • Moreover, the mere presence of an α-1,2-mannosidase in the cell does not, by itself, ensure proper intracellular trimming of Man8GlcNAc2 to Man5GlcNAc2. (See, e.g., Contreras et al. WO 02/00856 A2, in which an HDEL tagged mannosidase of T. reesei is localized primarily in the ER and co-expressed with an influenza haemagglutinin (HA) reporter protein on which virtually no Man5GlcNAc2 could be detected. See also Chiba et al. (1998) J. Biol. Chem. 273(41): 26298-26304, in which a chimeric α-1,2-mannosidase/Och1p transmembrane domain fusion localized in the ER, early Golgi and cytosol of S. cerevisiae, had no mannosidase trimming activity). Accordingly, mere localization of a mannosidase in the ER or Golgi is insufficient to ensure activity of the respective enzyme in that targeted organelle. (See also, Martinet et al. (1998) Biotech. Letters 20(12): 1171-1177, showing that α-1,2-mannosidase from T. reesei, while localizing intracellularly, increased rather than decreased the extent of mannosylation). To date, there is no report that demonstrates the intracellular localization of an active heterologous α-1,2- mannosidase in either yeast or fungi using a transmembrane localization sequence.
  • While it is useful to engineer strains that are able to produce Man5GlcNAc2 as the primary N-glycan structure, any attempt to further modify these high mannose precursor structures to more closely resemble human glycans requires additional in vivo or in vitro steps. Methods to further humanize glycans from fungal and yeast sources in vitro are described in U.S. Pat. No. 5,834,251 (supra). If Man5GlcNAc2 is to be further humanized in vivo, one has to ensure that the generated Man5GlcNAc2 structures are, in fact, generated intracellularly and not the product of mannosidase activity in the medium. Complex N-glycan formation in yeast or fungi will require high levels of Man5GlcNAc2 to be generated within the cell because only intracellular Man5GlcNAc2 glycans can be further processed to hybrid and complex N-glycans in vivo. In addition, one has to demonstrate that the majority of Man5GlcNAc2 structures generated are in fact a substrate for GnTI and thus allow the formation of hybrid and complex N-glycans.
  • Accordingly, the need exists for methods to produce glycoproteins characterized by a high intracellular Man5GlcNAc2 content which can be further processed into human-like glycoprotein structures in non-human eukaryotic host cells, and particularly in yeast and filamentous fungi.
  • N-Acetylglucosaminyltransferases
  • N-Acetylglucosaminyltransferases (“GnTs”) belong to another class of glycosylation enzymes that modify N-linked oligosaccharides in the secretory pathway. Such glycosyltransferases catalyze the transfer of a monosaccharide from specific sugar nucleotide donors onto particular hydroxyl position of a monosaccharide in a growing glycan chain in one of two possible anomeric linkages (either α or β). Dennis et al. (1999) Bioessays 21(5):412-21. Specific GnTs add N-acetylglucosamine (“GlcNAc”) onto the Manα1,6 arm or the Manα1,3 arm of an N-glycan substrate (e.g., Man5GlcNAc2 (“mannose-5 core”) and Man3GlcNAc2 (an “inner core structure”)). The reaction product (e.g., GlcNAcMan5GlcNAc2 or GlcNAc2Man3GlcNAc2) can then be modified into bi-, tri-, and tetra-antennary N-linked oligosaccharide structures.
  • N-Acetylglucosaminyltransferase III (“GnTIII”) is an enzyme that catalyzes the addition of a GlcNAc, on the middle mannose of the trimannose core (Manα1,6 (Manα1,3) Man β1,4-GlcNAc β1,4-GlcNAc β1,4-Asn) of an N-linked oligosaccharide. The addition by GnTIII of a bisecting GlcNAc to an acceptor substrate (e.g. trimannose core) yields a so-called bisected N-glycan. For example, the addition by GnTIII of a bisecting GlcNAc to the GlcNAcMan3GlcNAc2 structure may yield a bisected N-glycan, GlcNAc2Man3GlcNAc2. Similarly, the addition by GnTIII of a bisecting GlcNAc to a GlcNAc2Man3GlcNAc2 structure yields another bisected N-glycan, GlcNAc3Man3GlcNAc2. This latter structure has been implicated in greater antibody-dependent cellular cytotoxicity (ADCC). Umana et al. (1999) Nat. Biotechnol. 17(2): 176-80. Other bisected N-glycans can be formed by the action of GnTIII. For example, GlcNAcMan4GlcNAc2 can be converted to bisected GlcNAc2Man4GlcNAC2, Man5GlcNAc2 can be converted to bisected GlcNAcMan5GlcNAc2, and GlcNAcMan5GlcNAc2 can be converted to bisected GlcNAc2Man5GlcNAc2. See, e.g., Narasimhan (1982) J. Biol. Chem. 257:10235-42. Thus far, GnTIII activity has only been shown in mammalian cells.
  • Re-engineering glycoforms of immunoglobulins expressed by mammalian cells is a tedious and cumbersome task. Especially in the case of GnTIII, where over-expression of this enzyme has been implicated in growth inhibition, methods involving regulated (inducible) gene expression had to be employed to produce immunoglobulins with bisected N-glycans. Umana et al. (1999) Biotechnol Bioeng. 65(5):542-9; Umana et al. (1999) Nat. Biotechnol. 17(2):176-80; Umana et al WO 03/011878; U.S. Pat. No. 6,602,684. Such a growth-inhibition effect complicates the ability to coexpress the target protein and GnTIII and may impose an upper limit on GnTIII overexpression. U.S. Pat. No. 6,602,684. Careful optimization of the expression levels of GnTIII may be necessary. Id. What is needed, therefore, is a protein production system utilizing the inherent capability of robust product titers such as those produced in lower eukaryotic host cells (e.g., yeast and filamentous fungi), which is capable of producing bisected N-glycans on proteins, especially therapeutic proteins, expressed in these cells. As described above, however, development of the lower eukaryotic host cells used in such a protein production system requires that the endogenous glycosylation pathways of the host cells be further modified.
  • SUMMARY OF THE INVENTION
  • Host cells and cell lines having genetically modified glycosylation pathways that allow them to carry out a sequence of enzymatic reactions which mimic the processing of glycoproteins in mammals, especially in humans, have been developed. Recombinant proteins expressed in these engineered hosts yield glycoproteins more similar, if not substantially identical, to their mammalian, e.g., human counterparts. Host cells of the invention, e.g., lower eukaryotic microorganisms and other non-human, eukaryotic host cells grown in culture, are modified to produce N-glycans, such as bisected N-glycans, or other structures produced along human glycosylation pathways. This result is achieved using a combination of engineering and/or selection of strains that do not, for example, express enzymes that create the undesirable structures characteristic of the fungal glycoproteins and that do, for example, express heterologous enzymes capable of producing a “human-like” glycoprotein.
  • The present invention thus provides a glycoprotein production method using (1) a lower eukaryotic host such as a unicellular or filamentous fungus, or (2) any non-human eukaryotic organism that has a different glycosylation pattern from humans, to modify the glycosylation composition and structures of the proteins made in a host organism (“host cell”) so that they resemble more closely carbohydrate structures found in mammalian, e.g., human proteins. The process allows one to obtain an engineered host cell which can be used to express and target any desirable gene(s), e.g., one involved in glycosylation, by methods that are well-established in the scientific literature and generally known to the artisan in the field of protein expression. Host cells with modified oligosaccharides are created or selected. For the production of therapeutic proteins, this method may be adapted to engineer cell lines in which any desired glycosylation structure may be obtained.
  • Accordingly, in one embodiment, the invention provides methods for making a human-like glycoprotein in a lower eukaryotic host cell by introduction into the cell of an N-acetylglucosaminyltransferase III activity. In a preferred embodiment, the N-acetylglucosaminyltransferase III activity is expressed in the cell, and in an even more preferred embodiment, this expression results in the production of N-glycans comprising GlcNAc3Man3GlcNAc2, GlcNAc2Man3GlcNAc2, or GlcNAc2Man5GlcNAc2 bisected structures. In another preferred embodiment, the N-acetylglucosaminyltransferase III activity is substantially intracellular. In another preferred embodiment of the invention, the glycoprotein including the N-glycans with bisected structures is isolated from the lower eukaryotic host cell. In an even more preferred embodiment, the glycoprotein produced in the host cell is a therapeutic protein.
  • In another aspect, the invention provides a lower eukaryotic host cell that includes both an N-acetylglucosaminyltransferase III activity and an N-acetylglucosaminyltransferase II activity. In a preferred embodiment, the host cell including the N-acetylglucosaminyltransferase III activity produces N-glycans comprising GlcNAcMan3GlcNAc2 structures that are capable of reacting with this activity. In a more preferred embodiment, the activity produces a bisected glycan. The lower eukaryotic host cell of some embodiments of the invention may thus include an N-glycan with a bisected glycan. In a preferred embodiment, the N-glycan includes greater than 10 mole % of the bisected glycan. In some embodiments, the host cell includes an N-glycan that comprises GlcNAc3Man3GlcNAc2, GlcNAc2Man3GlcNAc2, or GlcNAc2Man5GlcNAc2 bisected structures. In a preferred embodiment, the host cell includes a Man5GlcNAc2 core structure or a Man3GlcNAc2 core structure that is modified by a bisecting GlcNAc. In an even more preferred embodiment, the cell produces greater than 10 mole % of the modified structure.
  • In another embodiment of the invention, the lower eukaryotic host cell contains an N-acetylglucosaminyltransferase I activity in addition to the N-acetylglucosaminyltransferase III activity. In a preferred embodiment, the activities are substantially intracellular. In another preferred embodiment, the cell produces N-glycans comprising GlcNAcMan3GlcNAc2 that are capable of reacting with the GnTIII activity. In an even more preferred embodiment, the GnTIII activity of the cell produces a bisected glycan.
  • In another embodiment, the lower eukaryotic host cell of the invention contains both an N-acetylglucosaminyltransferase III activity and a mannosidase II activity. In a preferred embodiment, the host cell further contains an N-acetylglucosaminyltransferase I activity. In another preferred embodiment, the host cell further contains an N-acetylglucosaminyltransferase II activity. In another preferred embodiment, the host cell further contains both an N-acetylglucosaminyltransferase I activity and an N-acetylglucosaminyltransferase II activity.
  • In another embodiment, the host cell of the invention is deficient in an OCH1 mannosyltransferase activity. Such a cell may, for example, be deficient in a Do1-P-Man:Man5GlcNAc2-PP-Do1 mannosyltransferase activity. In yet another embodiment, the host cell of the invention may further comprise an α-1,2-mannosidase I activity. In another embodiment, the host cell may further comprise a UDP-GlcNAc transporter.
  • The present invention also provides glycoproteins that are made by the processes of the invention. In one embodiment, the glycoprotein includes a bisecting GlcNAc on a Man5GlcNAc2 or a Man3GlcNAc2 core structure and is produced in a lower eukaryotic host cell. In another embodiment, the glycoprotein includes a bisecting GlcNAc attached to a Man5GlcNAc2, Man4GlcNAc2. Man3GlcNAc2, GlcNAcMan3GlcNAc2, GlcNAcMan5GlcNAc2, or a GlcNAc2Man3GlcNAc2 core structure and is produced in a lower eukaryotic host cell. In a preferred embodiment, greater than 10 mole % of the core structures of the glycoprotein of the invention are modified by the bisecting GlcNAc.
  • In another aspect, the invention provides pharmaceutical compositions that contain the human-like glycoproteins produced in a lower eukaryotic host cell. Also provided according to the invention are vectors encoding proteins having N-acetylglucosaminyltransferase III activity and containing attached targeting peptide sequences. In a preferred embodiment, the proteins encoded by the vectors are localized in a lower eukaryotic host cell such that they produce N-glycans having bisected structures.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A is a schematic diagram of a typical fungal N-glycosylation pathway.
  • FIG. 1B is a schematic diagram of a typical human N-glycosylation pathway.
  • FIG. 2 depicts construction of a combinatorial DNA library of fusion constructs. FIG. 2A diagrams the insertion of a targeting peptide fragment into pCR2.1-TOPO (Invitrogen, Carlsbad, Calif.). FIG. 2B shows the generated targeting peptide sub-library having restriction sites NotI-AscI. FIG. 2C diagrams the insertion of a catalytic domain region into pJN347, a modified pUC19 vector. FIG. 2D shows the generated catalytic domain sub-library having restriction sites NotI, AscI and PacI. FIG. 2E depicts one particular fusion construct generated from the targeting peptide sub-library and the catalytic domain sub-library.
  • FIG. 3 illustrates the M. musculus α-1,2-mannosidase IA open reading frame nucleic acid sequence (SEQ ID NO:48) and encoded polypeptide sequence (SEQ ID NO:49). The sequences of the PCR primers used to generate N-terminal truncations are underlined.
  • FIG. 4 illustrates engineering of vectors with multiple auxotrophic markers and genetic integration of target proteins in the P. pastoris OCH1 locus.
  • FIGS. 5A-5E show MALDI-TOF analysis demonstrating production of kringle 3 domain of human plasminogen (K3) glycoproteins having Man5GlcNAc2 as the predominant N-glycan structure in P. pastoris. FIG. 5A depicts the standard Man5GlcNAc2[a] glycan (Glyko, Novato, Calif.) and Man5GlcNAc2+Na+[b]. FIG. 5B shows PNGase—released glycans from K3 wild type. The N-glycans shown are as follows: Man9GlcNAc2[d]; Man10GlcNAc2[e]; Man11GlcNAc2[f]; Man12GlcNAc2[g]. FIG. 5C depicts the och1 deletion resulting in the production of Man8GlcNAc2[c] as the predominant N-glycan. FIGS. 5D and 5E show the production of Man5GlcNAc2[b] after in vivo trimming of Man8GlcNAc2 with a chimeric α-1,2-mannosidase. The predominant N-glycan is indicated by a peak with a mass (m/z) of 1253 consistent with its identification as Man5GlcNAc2[b].
  • FIGS. 6A-6F show MALDI-TOF analysis demonstrating production of IFN-β glycoproteins having Man5GlcNAc2 as the predominant N-glycan structure in P. pastoris. FIG. 6A shows the standard Man5GlcNAc2[a] and Man5GlcNAc2+Na+[b] as the standard (Glyko, Novato, Calif.). FIG. 6B shows PNGase—released glycans from IFN-β wildtype. FIG. 6C depicts the och1 knock-out producing Man8GlcNAc2[c]; Man9GlcNAc2[d]; Man10GlcNAc2[e]; Man11GlcNAc2[f]; Man12GlcNAc2[g]; and no production of Man5GlcNAc2[b]. FIG. 6D shows relatively small amount of Man5GlcNAc2[b] among other intermediate N-glycans Man8GlcNAc2[c] to Man12GlcNAc2[g]. FIG. 6E shows a significant amount of Man5GlcNAc2[b] relative to the other glycans Man8GlcNAc2[c] and Man9GlcNAc2[d] produced by pGC5 (Saccharomyces MNS1(m)/mouse mannosidase IB Δ99). FIG. 6F shows predominant production of Man5GlcNAc2[b] on the secreted glycoprotein IFN-β by pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187). The N-glycan is indicated by a peak with a mass (m/z) of 1254 consistent with its identification as Man5GlcNAc2[b].
  • FIG. 7 shows a high performance liquid chromatogram for: (A) Man9GlcNAc2 standard labeled with 2-AB (negative control); (B) supernatant of medium P. pastoris, Δoch1 transformed with pFB8 mannosidase, which demonstrates a lack of extracellular mannosidase activity in the supernatant; and (C) Man9GlcNAc2 standard labeled with 2-AB after exposure to T. reesei mannosidase (positive control).
  • FIG. 8 shows a high performance liquid chromatogram for: (A) Man9GlcNAc2 standard labeled with 2-AB (negative control); (B) supernatant of medium P. pastoris, Δoch1 transformed with pGC5 mannosidase, which demonstrates a lack of extracellular mannosidase activity in the supernatant; and (C) Man9GlcNAc2 standard labeled with 2-AB after exposure to T. reesei mannosidase (positive control).
  • FIG. 9 shows a high performance liquid chromatogram for: (A) Man9GlcNAc2 standard labeled with 2-AB (negative control); (B) supernatant of medium P. pastoris, Δoch1 transformed with pBC18-5 mannosidase, which demonstrates lack of extracellular mannosidase activity in the supernatant; and (C) supernatant of medium P. pastoris, Δoch1 transformed with pDD28-3, which demonstrates activity in the supernatant (positive control).
  • FIGS. 10A-10B demonstrate the activity of an UDP-GlcNAc transporter in the production of GlcNAcMan5GlcNAc2 in P. pastoris. FIG. 10A depicts a P. pastoris strain (YSH-3) with a human GnTI but without the UDP-GlcNAc transporter resulting in some production of GlcNAcMan5GlcNAc2[b] but a predominant production of Man5GlcNAc2[a]. FIG. 10B depicts the addition of UDP-GlcNAc transporter from K. lactis in a strain (PBP-3) with the human GnTI, which resulted in the predominant production of GlcNAcMan5GlcNAc2[b]. The single prominent peak of mass (m/z) at 1457 is consistent with its identification as GlcNAcMan5GlcNAc2[b] as shown in FIG. 10B.
  • FIG. 11 shows a pH optimum of a heterologous mannosidase enzyme encoded by pBB27-2 (Saccharomyces MNN10 (s)/C. elegans mannosidase IB Δ31) expressed in P. pastoris.
  • FIGS. 12A-12C show MALDI-TOF analysis of N-glycans released from a cell free extract of K. lactis. FIG. 12A shows the N-glycans released from wild-type cells, which includes high-mannose type N-glycans. FIG. 12B shows the N-glycans released from och1 mnn1 deleted cells, revealing a distinct peak of mass (m/z) at 1908 consistent with its identification as Man9GlcNAc2[d]. FIG. 12C shows the N-glycans released from ochl mnnl deleted cells after in vitro α-1,2-mannosidase digest corresponding to a peak consistent with Man5GlcNAc2.
  • FIG. 13 is a schematic of the structure of the dolichyl pyrophosphate-linked oligosaccharide.
  • FIG. 14 is a schematic of the generation of GlcNAc2Man3GlcNAc2 N-glycans from fungal host cells which are deficient in alg3, alg9, or alg12 activities.
  • FIG. 15 is a schematic of processing reactions required to produce mammalian-type oligosaccharide structures in a fungal host cell with an alg3, och1 genotype.
  • FIG. 16 shows S. cerevisiae Alg3 Sequence Comparisons (Blast) (SEQ ID NOs:9-20, respectively, in order of appearance)
  • FIG. 17 shows S. cerevisiae ALG3 (SEQ ID NO:21) and Alg3p (SEQ ID NO:22) Sequences
  • FIG. 18 shows P. pastoris ALG3 (SEQ ID NO:23) and Alg3p (SEQ ID NO:24) Sequences
  • FIG. 19 shows P. pastoris ALG3 Sequence Comparisons (Blast) (SEQ ID NOs:23-31, respectively, in order of appearance)
  • FIG. 20 shows K. lactis ALG3 (SEQ ID NO:33) and Alg3p (SEQ ID NO:34) Sequences
  • FIG. 21 shows K. lactis ALG3 Sequence Comparisons (Blast) (SEQ ID NOs:35-40, respectively, in order of appearance)
  • FIG. 22 shows a model of an IgG immunoglobulin. Heavy chain and light chain can be, based on similar secondary and tertiary structure, subdivided into domains. The two heavy chains (domains VH, C H1, C H2 and CH3) are linked through three disulfide bridges. The light chains (domains VL and CL) are linked by another disulfide bridge to the C H1 portion of the heavy chain and, together with the C H1 and VH fragments, make up the Fab region. Antigens bind to the terminal portion of the Fab region. Effector-functions, such as Fc-gamma-Receptor binding have been localized to the C H2 domain, just downstream of the hinge region and are influenced by N-glycosylation of asparagine 297 in the heavy chain.
  • FIG. 23 is a schematic overview of a modular IgG1 expression vector.
  • FIG. 24 shows M. musculis GnTIII Nucleic Acid (SEQ ID NO:45) And Amino Acid (SEQ ID NO:46) Sequences
  • FIG. 25 (top) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in a P. pastoris YSH-1 displaying a predominant peak at 1461 m/z corresponding to the the mass of GlcNAcMan5GlcNAc2 [d]; FIG. 25 (bottom) shows a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in a P. pastoris YSH-1 transformed with D. melanogaster mannosidase IIΔ74/S. cerevisiae MNN2(s) showing a predominant peak at 1140 m/z corresponding to the mass of GlcNAcMan3GlcNAc2[b] and other peaks corresponding to GlcNAcMan4GlcNAc2[c] at 1303 m/z and GlcNAcMan5GlcNAc2[d] at 1465 m/z. This strain was designated YSH-37.
  • FIG. 26 (top) is the MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in P. pastoris YSH-1 as shown in FIG. 25 (top); FIG. 26 (bottom) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-1 cells transformed with a pVA53 construct (S. cerevisiae MNN2(s)/mGnTIII). The peak at 1463 m/z corresponds the mass of GlcNAcMan5GlcNAc2 [d] and the peak at 1666 m/z corresponds to the mass of GlcNAc2Man5GlcNAc2[a].
  • FIG. 27 (top) is the MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in P. pastoris YSH-1 as shown in FIG. 25 (top); FIG. 27 (bottom) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-1 cells transformed with a pVA55 construct (S. cerevisiae MNN2(s)/mGnTIII). The peak at 1463 m/z corresponds to the mass of GlcNAcMan5GlcNAc2[d] and the peak at 1667 m/z corresponds to the mass of GlcNAc2Man5GlcNAc2[a].
  • FIG. 28 (top) is the MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein produced in P. pastoris YSH-1 as shown in FIG. 25 (top); FIG. 28 (bottom) is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-1 cells transformed with a pVB51 construct (K. lactis GNT1(s)/mGnTIII). The predominant peak at 1463 m/z corresponds to the mass of GlcNAcMan5GlcNAc2[d] and a second peak at 1726 m/z [e], which does not correpond to the mass of GlcNAc2Man5GlcNAc2 is observed.
  • FIG. 29 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-44 cells. The predominant peak at 1356 m/z corresponds to the mass of GlcNAc2Man3GlcNAc2[x].
  • FIG. 30 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris YSH-44 cells transformed with a pVA53 construct (S. cerevisiae MAN2(s)/mGnTIII). The peak at 1340 m/z corresponds to the mass of GlcNAc2Man3GlcNAc2 [x] and the peak at 1542 m/z corresponds to the mass of GlcNAc3Man3GlcNAc2[y].
  • FIG. 31 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris PBP6-5 cells. The predominant peak at 1340 m/z corresponds to the mass of GlcNAc2Man3GlcNAc2 [X].
  • FIG. 32 is a MALDI-TOF-MS analysis of N-glycans isolated from a kringle 3 glycoprotein expressed in P. pastoris PBP6-5 cells transformed with a pVA53 construct (S. cerevisiae MNN2(s)/mGnTIII). The peak at 1340 m/z corresponds to the mass of GlcNAc2Man3GlcNAc2[x] and the peak at 1543 m/z correponds to the mass of GlcNAc3Man3GlcNAc2[y].
  • FIG. 33 shows a high performance liquid chromatogram, which demonstrates a lack of extracellular GnTIII activity (pVA53) in the supernatant. The N-glycan GlcNAcMan5GlcNAc2 purified from K3 expressed in PBP-3 strain was added to: BMMY (A); 1 mM UDP-GlcNAc (Sigma Chemical Co., St. Louis, Mo.)) in BMMY (B); the supernatant of YSH-44 transformed with pVA53 [YSH-57] (C); and the supernatant of YSH-57+1 mM UDP-GlcNAc (D).
  • FIG. 34 shows a high performance liquid chromatogram, which demonstrates a lack of extracellular GnTIII activity (pVA53) in the supernatant. The N-glycan GlcNAc2Man3GlcNAc2 purified from K3 expressed in YSH-44 strain was added to: BMMY (A); 1 mM UDP-GlcNAc (Sigma Chemical Co., St. Louis, Mo.)) in BMMY (B); and the supernatant of YSH-44 transformed with pVA53 [YSH-57] (C).
  • FIG. 35 is a schematic diagram comparing the normal glycosylation pathways in humans and P. pastoris (Panel A) with an engineered humanized N-glycosylation pathway in lower eukaryotes (Panel B). The engineered pathway represents the construction of P. pastoris strain PBP6-5, which after modification with GnTIII becomes P. pastoris strain PBP38.
  • FIG. 36 is a schematic diagram showing the predominant secreted glycoform produced by each of the designated P. pastoris strains and the gene modification used to engineer each of the strains.
  • FIG. 37 is a structural representation of the transfer of a GlcNAc to the oligosaccharide intermediate, GlcNAcMan5GlcNAc2, produced on glycoproteins in a lower eukaryotic host cell, as catalyzed by GnTIII.
  • FIG. 38 is a structural representation of the transfer of a GlcNAc to the oligosaccharide intermediate, GlcNAcMan3GlcNAc2, produced on glycoproteins in a lower eukaryotic host cell, as catalyzed by GnTII, and the subsequent transfer of a GlcNAc to the product of that reaction, GlcNAc2Man3GlcNAc2, as catalyzed by GnTIII.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. The methods and techniques of the present invention are generally performed according to conventional methods well known in the art. Generally, nomenclatures used in connection with, and techniques of biochemistry, enzymology, molecular and cellular biology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well-known and commonly used in the art.
  • The methods and techniques of the present invention are generally performed according to conventional methods well-known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification unless otherwise indicated. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989); Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates (1992, and Supplements to 2002); Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1990); Introduction to Glycobiology, Maureen E. Taylor, Kurt Drickamer, Oxford Univ. Press (2003); Worthington Enzyme Manual, Worthington Biochemical Corp. Freehold, N.J.; Handbook of Biochemistry: Section A Proteins, Vol I 1976 CRC Press; Handbook of Biochemistry: Section A Proteins, Vol II 1976 CRC Press; Essentials of Glycobiology, Cold Spring Harbor Laboratory Press (1999). The nomenclatures used in connection with, and the laboratory procedures and techniques of, molecular and cellular biology, protein biochemistry, enzymology and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art.
  • All publications, patents and other references mentioned herein are incorporated by reference.
  • The following terms, unless otherwise indicated, shall be understood to have the following meanings:
  • As used herein, the term “N-glycan” refers to an N-linked oligosaccharide, e.g., one that is attached by an asparagine-N-acetylglucosamine linkage to an asparagine residue of a polypeptide. N-glycans have a common pentasaccharide core of Man3GlcNAc2 (“Man” refers to mannose; “Glc” refers to glucose; and “NAc” refers to N-acetyl; GlcNAc refers to N-acetylglucosamine). The term “trimannose core” used with respect to the N-glycan also refers to the structure Man3GlcNAc2 (“Man3”). The term “pentamannose core” or “Mannose-5 core” or “Man5” used with respect to the N-glycan refers to the structure Man5GlcNAc2. N-glycans differ with respect to the number of branches (antennae) comprising peripheral sugars (e.g., GlcNAc, fucose, and sialic acid) that are attached to the Man3 core structure. N-glycans are classified according to their branched constituents (e.g., high mannose, complex or hybrid).
  • A “high mannose” type N-glycan has five or more mannose residues. A “complex” type N-glycan typically has at least one GlcNAc attached to the 1,3 mannose arm and at least one GlcNAc attached to the 1,6 mannose arm of the trimannose core. Complex N-glycans may also have galactose (“Gal”) residues that are optionally modified with sialic acid or derivatives (“NeuAc”, where “Neu” refers to neuraminic acid and “Ac” refers to acetyl). A complex N-glycan typically has at least one branch that terminates in an oligosaccharide such as, for example: NeuNAc-; NeuAcα2-6GalNAcα1-; NeuAcα2-3Galβ1-3GalNAcα1-; NeuAcα2-3/6Galβ1-4GlcNAcβ1-; GlcNAcα1-4Galβ1-(mucins only); Fucα1-2Galβ1-(blood group H). Sulfate esters can occur on galactose, GalNAc, and GlcNAc residues, and phosphate esters can occur on mannose residues. NeuAc (Neu: neuraminic acid; Ac:acetyl) can be O-acetylated or replaced by NeuG1 (N-glycolylneuraminic acid). Complex N-glycans may also have intrachain substitutions comprising “bisecting” GlcNAc and core fucose (“Fuc”). A “hybrid” N-glycan has at least one GlcNAc on the terminal of the 1,3 mannose arm of the trimannose core and zero or more mannoses on the 1,6 mannose arm of the trimannose core.
  • The term “predominant” or “predominantly” used with respect to the production of N-glycans refers to a structure which represents the major peak detected by matrix assisted laser desorption ionization time of flight mass spectrometry (MALDI-TOF) analysis.
  • Abbreviations used herein are of common usage in the art, see, e.g., abbreviations of sugars, above. Other common abbreviations include “PNGase”, which refers to peptide N-glycosidase F (EC 3.2.2.18); “GlcNAc Tr” or “GnT,” which refers to N-acetylglucosaminyl Transferase enzymes; “NANA” refers to N-acetylneuraminic acid.
  • As used herein, a “humanized glycoprotein” or a “human-like glycoprotein” refers alternatively to a protein having attached thereto N-glycans having fewer than four mannose residues, and synthetic glycoprotein intermediates (which are also useful and can be manipulated further in vitro or in vivo) having at least five mannose residues. Preferably, glycoproteins produced according to the invention contain at least 30 mole %, preferably at least 40 mole % and more preferably 50, 60, 70, 80, 90, or even 100 mole % of the Man5GlcNAc2 intermediate, at least transiently. This may be achieved, e.g., by engineering a host cell of the invention to express a “better”, i.e., a more efficient glycosylation enzyme. For example, a mannosidase is selected such that it will have optimal activity under the conditions present at the site in the host cell where proteins are glycosylated and is introduced into the host cell preferably by targeting the enzyme to a host cell organelle where activity is desired.
  • The term “enzyme”, when used herein in connection with altering host cell glycosylation, refers to a molecule having at least one enzymatic activity, and includes full-length enzymes, catalytically active fragments, chimerics, complexes, and the like. A “catalytically active fragment” of an enzyme refers to a polypeptide having a detectable level of functional (enzymatic) activity. Enzyme activity is “substantially intracellular” when less than 10% of the enzyme activity is measurable outside the cell compared to that measurable from lysed cells.
  • A lower eukaryotic host cell, when used herein in connection with glycosylation profiles, refers to any eukaryotic cell which ordinarily produces high mannose containing N-glycans, and thus is meant to include some animal or plant cells and most typical lower eukaryotic cells, including uni- and multicellular fungal and algal cells.
  • As used herein, the term “secretion pathway” refers to the assembly line of various glycosylation enzymes to which a lipid-linked oligosaccharide precursor and an N-glycan substrate are sequentially exposed, following the molecular flow of a nascent polypeptide chain from the cytoplasm to the endoplasmic reticulum (ER) and the compartments of the Golgi apparatus. Enzymes are said to be localized along this pathway. An enzyme X that acts on a lipid-linked glycan or an N-glycan before enzyme Y is said to be or to act “upstream” to enzyme Y; similarly, enzyme Y is or acts “downstream” from enzyme X.
  • The term “targeting peptide” as used herein refers to nucleotide or amino acid sequences encoding a cellular targeting signal peptide which mediates the localization. (or retention) of an associated sequence to sub-cellular locations, e.g., organelles.
  • The term “polynucleotide” or “nucleic acid molecule” refers to a polymeric form of nucleotides of at least 10 bases in length. The term includes DNA molecules (e.g., cDNA or genomic or synthetic DNA) and RNA molecules (e.g., mRNA or synthetic RNA), as well as analogs of DNA or RNA containing non-natural nucleotide analogs, non-native internucleoside bonds, or both. The nucleic acid can be in any topological conformation. For instance, the nucleic acid can be single-stranded, double-stranded, triple-stranded, quadruplexed, partially double-stranded, branched, hairpinned, circular, or in a padlocked conformation. The term includes single and double stranded forms of DNA. A nucleic acid molecule of this invention may include both sense and antisense strands of RNA, cDNA, genomic DNA, and synthetic forms and mixed polymers of the above. They may be modified chemically or biochemically or may contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those of skill in the art. Such modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen, etc.), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids, etc.) Also included are synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen bonding and other chemical interactions. Such molecules are known in the art and include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
  • Unless otherwise indicated, a “nucleic acid comprising SEQ ID NO:X” refers to a nucleic acid, at least a portion of which has either (i) the sequence of SEQ ID NO:X, or (ii) a sequence complementary to SEQ ID NO:X. The choice between the two is dictated by the context. For instance, if the nucleic acid is used as a probe, the choice between the two is dictated by the requirement that the probe be complementary to the desired target.
  • An “isolated” or “substantially pure” nucleic acid or polynucleotide (e.g., an RNA, DNA or a mixed polymer) is one which is substantially separated from other cellular components that naturally accompany the native polynucleotide in its natural host cell, e.g., ribosomes, polymerases, and genomic sequences with which it is naturally associated. The term embraces a nucleic acid or polynucleotide that (1) has been removed from its naturally occurring environment, (2) is not associated with all or a portion of a polynucleotide in which the “isolated polynucleotide” is found in nature, (3) is operatively linked to a polynucleotide which it is not linked to in nature, or (4) does not occur in nature. The term “isolated” or “substantially pure” also can be used in reference to recombinant or cloned DNA isolates, chemically synthesized polynucleotide analogs, or polynucleotide analogs that are biologically synthesized by heterologous systems.
  • However, “isolated” does not necessarily require that the nucleic acid or polynucleotide so described has itself been physically removed from its native environment. For instance, an endogenous nucleic acid sequence in the genome of an organism is deemed “isolated” herein if a heterologous sequence (i.e., a sequence that is not naturally adjacent to this endogenous nucleic acid sequence) is placed adjacent to the endogenous nucleic acid sequence, such that the expression of this endogenous nucleic acid sequence is altered. By way of example, a non-native promoter sequence can be substituted (e.g., by homologous recombination) for the native promoter of a gene in the genome of a human cell, such that this gene has an altered expression pattern. This gene would now become “isolated” because it is separated from at least some of the sequences that naturally flank it.
  • A nucleic acid is also considered “isolated” if it contains any modifications that do not naturally occur to the corresponding nucleic acid in a genome. For instance, an endogenous coding sequence is considered “isolated” if it contains an insertion, deletion or a point mutation introduced artificially, e.g., by human intervention. An “isolated nucleic acid” also includes a nucleic acid integrated into a host cell chromosome at a heterologous site, a nucleic acid construct present as an episome. Moreover, an “isolated nucleic acid” can be substantially free of other cellular material, or substantially free of culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • As used herein, the phrase “degenerate variant” of a reference nucleic acid sequence encompasses nucleic acid sequences that can be translated, according to the standard genetic code, to provide an amino acid sequence identical to that translated from the reference nucleic acid sequence.
  • The term “percent sequence identity” or “identical” in the context of nucleic acid sequences refers to the residues in the two sequences which are the same when aligned for maximum correspondence. The length of sequence identity comparison may be over a stretch of at least about nine nucleotides, usually at least about 20 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36 or more nucleotides. There are a number of different algorithms known in the art which can be used to measure nucleotide sequence identity. For instance, polynucleotide sequences can be compared using FASTA, Gap or Bestfit, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wis. FASTA provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson (1990) Methods Enzymol. 183:63-98, incorporated herein by reference in its entirety). For instance, percent sequence identity between nucleic acid sequences can be determined using FASTA with its default parameters (a word size of 6 and the NOPAM factor for the scoring matrix) or using Gap with its default parameters as provided in GCG Version 6.1, herein incorporated by reference.
  • The term “substantial homology” or “substantial similarity,” when referring to a nucleic acid or fragment thereof, indicates that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 50%, more preferably 60% of the nucleotide bases, usually at least about 70%, more usually at least about 80%, preferably at least about 90%, and more preferably at least about 95%, 96%, 97%, 98% or 99% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or Gap, as discussed above.
  • Alternatively, substantial homology or similarity exists when a nucleic acid or fragment thereof hybridizes to another nucleic acid, to a strand of another nucleic acid, or to the complementary strand thereof, under stringent hybridization conditions. “Stringent hybridization conditions” and “stringent wash conditions” in the context of nucleic acid hybridization experiments depend upon a number of different physical parameters. Nucleic acid hybridization will be affected by such conditions as salt concentration, temperature, solvents, the base composition of the hybridizing species, length of the complementary regions, and the number of nucleotide base mismatches between the hybridizing nucleic acids, as will be readily appreciated by those skilled in the art. One having ordinary skill in the art knows how to vary these parameters to achieve a particular stringency of hybridization.
  • In general, “stringent hybridization” is performed at about 25° C. below the thermal melting point (Tm) for the specific DNA hybrid under a particular set of conditions. “Stringent washing” is performed at temperatures about 5° C. lower than the Tm for the specific DNA hybrid under a particular set of conditions. The Tm is the temperature at which 50% of the target sequence hybridizes to a perfectly matched probe. See Sambrook et al., supra, page 9.51, hereby incorporated by reference. For purposes herein, “high stringency conditions” are defined for solution phase hybridization as aqueous hybridization (i.e., free of formamide) in 6×SSC (where 20×SSC contains 3.0 M NaCl and 0.3 M sodium citrate), 1% SDS at 65° C. for 8-12 hours, followed by two washes in 0.2×SSC, 0.1% SDS at 65° C. for 20 minutes. It will be appreciated by the skilled artisan that hybridization at 65° C. will occur at different rates depending on a number of factors including the length and percent identity of the sequences which are hybridizing.
  • The term “mutated” when applied to nucleic acid sequences means that nucleotides in a nucleic acid sequence may be inserted, deleted or changed compared to a reference nucleic acid sequence. A single alteration may be made at a locus (a point mutation) or multiple nucleotides may be inserted, deleted or changed at a single locus. In addition, one or more alterations may be made at any number of loci within a nucleic acid sequence. A nucleic acid sequence may be mutated by any method known in the art including but not limited to mutagenesis techniques such as “error-prone PCR” (a process for performing PCR under conditions where the copying fidelity of the DNA polymerase is low, such that a high rate of point mutations is obtained along the entire length of the PCR product. See, e.g., Leung et al. (1989) Technique 1:11-15 and Caldwell and Joyce (1992) PCR Methods Applic. 2:28-33); and “oligonucleotide-directed mutagenesis” (a process which enables the generation of site-specific mutations in any cloned DNA segment of interest. See, e.g., Reidhaar-Olson et al. (1988) Science 241:53-57).
  • The term “vector” as used herein is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Other vectors include cosmids, bacterial artificial chromosomes (BAC) and yeast artificial chromosomes (YAC). Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome (discussed in more detail below). Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., vectors having an origin of replication which functions in the host cell). Other vectors can be integrated into the genome of a host cell upon introduction into the host cell, and are thereby replicated along with the host genome. Moreover, certain preferred vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
  • “Operatively linked” expression control sequences refers to a linkage in which the expression control sequence is contiguous with the gene of interest to control the gene of interest, as well as expression control sequences that act in trans or at a distance to control the gene of interest.
  • The term “expression control sequence” as used herein refers to polynucleotide sequences which are necessary to affect the expression of coding sequences to which they are operatively linked. Expression control sequences are sequences which control the transcription, post-transcriptional events and translation of nucleic acid sequences. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (e.g., ribosome binding sites); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence. The term “control sequences” is intended to include, at a minimum, all components whose presence is essential for expression, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • The term “recombinant host cell” (or simply “host cell”), as used herein, is intended to refer to a cell into which a nucleic acid such as a recombinant vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein. A recombinant host cell may be an isolated cell or cell line grown in culture or may be a cell which resides in a living tissue or organism.
  • The term “peptide” as used herein refers to a short polypeptide, e.g., one that is typically less than about 50 amino acids long and more typically less than about 30 amino acids long. The term as used herein encompasses analogs and mimetics that mimic structural and thus biological function.
  • The term “polypeptide” as used herein encompasses both naturally-occurring and non-naturally-occurring proteins, and fragments, mutants, derivatives and analogs thereof. A polypeptide may be monomeric or polymeric. Further, a polypeptide may comprise a number of different domains each of which has one or more distinct activities.
  • The term “isolated protein” or “isolated polypeptide” is a protein or polypeptide that by virtue of its origin or source of derivation (1) is not associated with naturally associated components that accompany it in its native state, (2) when it exists in a purity not found in nature, where purity can be adjudged with respect to the presence of other cellular material (e.g., is free of other proteins from the same species) (3) is expressed by a cell from a different species, or (4) does not occur in nature (e.g., it is a fragment of a polypeptide found in nature or it includes amino acid analogs or derivatives not found in nature or linkages other than standard peptide bonds). Thus, a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components. A polypeptide or protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well-known in the art. As thus defined, “isolated” does not necessarily require that the protein, polypeptide, peptide or oligopeptide so described has been physically removed from its native environment.
  • The term “polypeptide fragment” as used herein refers to a polypeptide that has an amino-terminal and/or carboxy-terminal deletion compared to a full-length polypeptide. In a preferred embodiment, the polypeptide fragment is a contiguous sequence in which the amino acid sequence of the fragment is identical to the corresponding positions in the naturally-occurring sequence. Fragments typically are at least 5, 6, 7, 8, 9 or 10 amino acids long, preferably at least 12, 14, 16 or 18 amino acids long, more preferably at least 20 amino acids long, more preferably at least 25, 30, 35, 40 or 45, amino acids, even more preferably at least 50 or 60 amino acids long, and even more preferably at least 70 amino acids long.
  • A “modified derivative” refers to polypeptides or fragments thereof that are substantially homologous in primary structural sequence but which include, e.g., in vivo or in vitro chemical and biochemical modifications or which incorporate amino acids that are not found in the native polypeptide. Such modifications include, for example, acetylation, carboxylation, phosphorylation, glycosylation, ubiquitination, labeling, e.g., with radionuclides, and various enzymatic modifications, as will be readily appreciated by those well skilled in the art. A variety of methods for labeling polypeptides and of substituents or labels useful for such purposes are well-known in the art, and include radioactive isotopes such as 125I, 32P, 35S, and 3H, ligands which bind to labeled antiligands (e.g., antibodies), fluorophores, chemiluminescent agents, enzymes, and antiligands which can serve as specific binding pair members for a labeled ligand. The choice of label depends on the sensitivity required, ease of conjugation with the primer, stability requirements, and available instrumentation. Methods for labeling polypeptides are well-known in the art. See Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates (1992, and Supplements to 2002), hereby incorporated by reference.
  • A “polypeptide mutant” or “mutein” refers to a polypeptide whose sequence contains an insertion, duplication, deletion, rearrangement or substitution of one or more amino acids compared to the amino acid sequence of a native or wild-type protein. A mutein may have one or more amino acid point substitutions, in which a single amino acid at a position has been changed to another amino acid, one or more insertions and/or deletions, in which one or more amino acids are inserted or deleted, respectively, in the sequence of the naturally-occurring protein, and/or truncations of the amino acid sequence at either or both the amino or carboxy termini. A mutein may have the same but preferably has a different biological activity compared to the naturally-occurring protein.
  • A mutein has at least 70% overall sequence homology to its wild-type counterpart. Even more preferred are muteins having 80%, 85% or 90% overall sequence homology to the wild-type protein. In an even more preferred embodiment, a mutein exhibits 95% sequence identity, even more preferably 97%, even more preferably 98% and even more preferably 99% overall sequence identity. Sequence homology may be measured by any common sequence analysis algorithm, such as Gap or Bestfit.
  • Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinity or enzymatic activity, and (5) confer or modify other physicochemical or functional properties of such analogs.
  • As used herein, the twenty conventional amino acids and their abbreviations follow conventional usage. See Immunology-A Synthesis (2nd Edition, E. S. Golub and D. R. Gren, Eds., Sinauer Associates, Sunderland, Mass. (1991)), which is incorporated herein by reference. Stereoisomers (e.g., D-amino acids) of the twenty conventional amino acids, unnatural amino acids such as α-, α-disubstituted amino acids, N-alkyl amino acids, and other unconventional amino acids may also be suitable components for polypeptides of the present invention. Examples of unconventional amino acids include: 4-hydroxyproline, γ-carboxyglutamate, ε-N,N,N-trimethyllysine, ε-N-acetyllysine, O-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, N-methylarginine, and other similar amino acids and imino acids (e.g., 4-hydroxyproline). In the polypeptide notation used herein, the left-hand direction is the amino terminal direction and the right hand direction is the carboxy-terminal direction, in accordance with standard usage and convention.
  • A protein has “homology” or is “homologous” to a second protein if the nucleic acid sequence that encodes the protein has a similar sequence to the nucleic acid sequence that encodes the second protein. Alternatively, a protein has homology to a second protein if the two proteins have “similar” amino acid sequences. (Thus, the term “homologous proteins” is defined to mean that the two proteins have similar amino acid sequences). In a preferred embodiment, a homologous protein is one that exhibits 60% sequence homology to the wild type protein, more preferred is 70% sequence homology. Even more preferred are homologous proteins that exhibit 80%, 85% or 90% sequence homology to the wild type protein. In a yet more preferred embodiment, a homologous protein exhibits 95%, 97%, 98% or 99% sequence identity. As used herein, homology between two regions of amino acid sequence (especially with respect to predicted structural similarities) is interpreted as implying similarity in function.
  • When “homologous” is used in reference to proteins or peptides, it is recognized that residue positions that are not identical often differ by conservative amino acid substitutions. A “conservative amino acid substitution” is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein. In cases where two or more amino acid sequences differ from each other by conservative substitutions, the percent sequence identity or degree of homology may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well known to those of skill in the art (see, e.g., Pearson (1990) Methods Enzymol. 183:63-98).
  • The following six groups each contain amino acids that are conservative substitutions for one another: 1) Serine (S), Threonine (T); 2) Aspartic Acid (D), Glutamic Acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Alanine (A), Valine (V), and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W).
  • Sequence homology for polypeptides, which is also referred to as percent sequence identity, is typically measured using sequence analysis software. See, e.g., the Sequence Analysis Software Package of the Genetics Computer Group (GCG), University of Wisconsin Biotechnology Center, 910 University Avenue, Madison, Wis. 53705. Protein analysis software matches similar sequences using measure of homology assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions. For instance, GCG contains programs such as “Gap” and “Bestfit” which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1.
  • A preferred algorithm when comparing a inhibitory molecule sequence to a database containing a large number of sequences from different organisms is the computer program BLAST (Altschul et al. (1990) J. Mol. Biol. 215:403-410; Gish and States (1993) Nature Genet. 3:266-272; Madden et al. (1996) Meth. Enzymol. 266:131-141; Altschul et al. (1997) Nucleic Acids Res.25:3389-3402; Zhang and Madden (1997) Genome Res. 7:649-656), especially blastp or tblastn (Altschul et al., 1997). Preferred parameters for BLASTp are: Expectation value: 10 (default); Filter: seg (default); Cost to open a gap: 11 (default); Cost to extend a gap: 1 (default); Max. alignments: 100 (default); Word size: 11 (default); No. of descriptions: 100 (default); Penalty Matrix: BLOWSUM62.
  • The length of polypeptide sequences compared for homology will generally be at least about 16 amino acid residues, usually at least about 20 residues, more usually at least about 24 residues, typically at least about 28 residues, and preferably more than about 35 residues. When searching a database containing sequences from a large number of different organisms, it is preferable to compare amino acid sequences. Database searching using amino acid sequences can be measured by algorithms other than blastp known in the art. For instance, polypeptide sequences can be compared using FASTA, a program in GCG Version 6.1. FASTA provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (see Pearson (1990) Methods Enzymol. 183:63-98). For example, percent sequence identity between amino acid sequences can be determined using FASTA with its default parameters (a word size of 2 and the PAM250 scoring matrix), as provided in GCG Version 6.1, herein incorporated by reference.
  • The term “fusion protein” refers to a polypeptide comprising a polypeptide or fragment coupled to heterologous amino acid sequences. Fusion proteins are useful because they can be constructed to contain two or more desired functional elements from two or more different proteins. A fusion protein comprises at least 10 contiguous amino acids from a polypeptide of interest, more preferably at least 20 or 30 amino acids, even more preferably at least 40, 50 or 60 amino acids, yet more preferably at least 75, 100 or 125 amino acids. Fusion proteins can be produced recombinantly by constructing a nucleic acid sequence which encodes the polypeptide or a fragment thereof in-frame with a nucleic acid sequence encoding a different protein or peptide and then expressing the fusion protein. Alternatively, a fusion protein can be produced chemically by crosslinking the polypeptide or a fragment thereof to another protein.
  • The term “region” as used herein refers to a physically contiguous portion of the primary structure of a biomolecule. In the case of proteins, a region is defined by a contiguous portion of the amino acid sequence of that protein.
  • The term “domain” as used herein refers to a structure of a biomolecule that contributes to a known or suspected function of the biomolecule. Domains may be co-extensive with regions or portions thereof; domains may also include distinct, non-contiguous regions of a biomolecule. Examples of protein domains include, but are not limited to, an Ig domain, an extracellular domain, a transmembrane domain, and a cytoplasmic domain.
  • As used herein, the term “molecule” means any compound, including, but not limited to, a small molecule, peptide, protein, sugar, nucleotide, nucleic acid, lipid, etc., and such a compound can be natural or synthetic.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Exemplary methods and materials are described below, although methods and materials similar or equivalent to those described herein can also be used in the practice of the present invention and will be apparent to those of skill in the art. All publications and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. The materials, methods, and examples are illustrative only and not intended to be limiting.
  • Throughout this specification and claims, the word “comprise” or variations such as “comprises” or “comprising”, will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.
  • Methods For Producing Human-like Glycoproteins In Lower Eukaryotic Host Cells
  • The invention provides methods for producing a glycoprotein having human-like glycosylation in a non-human eukaryotic host cell. As described in more detail below, a eukaryotic host cell that does not naturally express, or which is engineered not to express, one or more enzymes involved in production of high mannose structures is selected as a starting host cell. Such a selected host cell is engineered to express one or more enzymes or other factors required to produce human-like glycoproteins. A desired host strain can be engineered one enzyme or more than one enzyme at a time. In addition, a nucleic acid molecule encoding one or more enzymes or activities may be used to engineer a host strain of the invention. Preferably, a library of nucleic acid molecules encoding potentially useful enzymes (e.g., chimeric enzymes comprising a catalytically active enzyme fragment ligated in-frame to a heterologous subcellular targeting sequence) is created (e.g., by ligation of sub-libraries comprising enzymatic fragments and subcellular targeting sequences), and a strain having one or more enzymes with optimal activities or producing the most “human-like” glycoproteins may be selected by transforming target host cells with one or more members of the library.
  • In particular, the methods described herein enable one to obtain, in vivo, Man5GlcNAc2 structures in high yield, at least transiently, for the purpose of further modifying it to yield complex N-glycans. A successful scheme to obtain suitable Man5GlcNAc2 structures in appropriate yields in a host cell, such as a lower eukaryotic organism, generally involves two parallel approaches: (1) reducing high mannose structures made by endogenous mannosyltransferase activities, if any, and (2) removing 1,2-α-mannose by mannosidases to yield high levels of suitable Man5GlcNAc2 structures which may be further reacted inside the host cell to form complex, human-like glycoforms.
  • Accordingly, a first step involves the selection or creation of a eukaryotic host cell, e.g., a lower eukaryote, capable of producing a specific precursor structure of Man5GlcNAc2 that is able to accept in vivo GlcNAc by the action of a GlcNAc transferase I (“GnTI”). In one embodiment, the method involves making or using a non-human eukaryotic host cell depleted in a 1,6 mannosyltransferase activity with respect to the N-glycan on a glycoprotein. Preferably, the host cell is depleted in an initiating 1,6 mannosyltransferase activity (see below). Such a host cell will lack one or more enzymes involved in the production of high mannose structures which are undesirable for producing human-like glycoproteins.
  • One or more enzyme activities are then introduced into such a host cell to produce N-glycans within the host cell characterized by having at least 30 mol % of the Man5GlcNAc2 (“Man5”) carbohydrate structures. Man5GlcNAc2 structures are necessary for complex N-glycan formation: Man5GlcNAc2 must be formed in vivo in a high yield (e.g., in excess of 30%), at least transiently, as subsequent mammalian- and human-like glycosylation reactions require Man5GlcNAc2 or a derivative thereof.
  • This step also requires the formation of a particular isomeric structure of Man5GlcNAc2 within the cell at a high yield. While Man5GlcNAc2 structures are necessary for complex N-glycan formation, their presence is by no means sufficient. That is because Man5GlcNAc2 may occur in different isomeric forms, which may or may not serve as a substrate for GlcNAc transferase I. As most glycosylation reactions are not complete, a particular glycosylated protein generally contains a range of different carbohydrate structures (i.e., glycoforms) on its surface. Thus, the mere presence of trace amounts (i.e., less than 5%) of a particular structure like Man5GlcNAc2 is of little practical relevance for producing mammalian- or human-like glycoproteins. It is the formation of a GlcNAc transferase I-accepting Man5GlcNAc2 intermediate (FIG. 1B) in high yield (i.e., above 30%), which is required. The formation of this intermediate is necessary to enable subsequent in vivo synthesis of complex N-glycans on glycosylated proteins of interest (target proteins).
  • Accordingly, some or all of the Man5GlcNAc2 produced by the selected host cell must be a productive substrate for enzyme activities along a mammalian glycosylation pathway, e.g., can serve as a substrate for a GlcNAc transferase I activity in vivo, thereby forming the human-like N-glycan intermediate GlcNAcMan5GlcNAc2 in the host cell. In a preferred embodiment, at least 10%, more preferably at least 30% and most preferably 50% or more of the Man5GlcNAc2 intermediate produced in the host cell of the invention is a productive substrate for GnTI in vivo. It is understood that if, for example, GlcNAcMan5GlcNAc2 is produced at 10% and Man5GlcNAc2 is produced at 25% on a target protein, that the total amount of transiently produced Man5GlcNAc2 is 35% because GlcNAcMan5GlcNAc2 is a product of Man5GlcNAc2.
  • One of ordinary skill in the art can select host cells from nature, e.g., existing fungi or other lower eukaryotes that produce significant levels of Man5GlcNAc2 in vivo. As yet, however, no lower eukaryote has been shown to provide such structures in vivo in excess of 1.8% of the total N-glycans (see e.g. Maras et al. (1997) Eur. J. Biochem. 249:701-707). Alternatively, such host cells may be genetically engineered to produce the Man5GlcNAc2 structure in vivo. Methods such as those described in U.S. Pat. No. 5,595,900 may be used to identify the absence or presence of particular glycosyltransferases, mannosidases and sugar nucleotide transporters in a target host cell or organism of interest.
  • Inactivation of Undesirable Host Cell Glycosylation Enzymes
  • The methods of the invention are directed to making host cells which produce glycoproteins having altered, and preferably human-like, N-glycan structures. In a preferred embodiment, the methods are directed to making host cells in which oligosaccharide precursors are enriched in Man5GlcNAc2. Preferably, a eukaryotic host cell is used that does not express one or more enzymes involved in the production of high mannose structures. Such a host cell may be found in nature or may be engineered, e.g., starting with or derived from one of many such mutants already described in yeasts. Thus, depending on the selected host cell, one or a number of genes that encode enzymes known to be characteristic of non-human glycosylation reactions will have to be deleted. Such genes and their corresponding proteins have been extensively characterized in a number of lower eukaryotes (e.g., S. cerevisiae, T. reesei, A. nidulans, etc.), thereby providing a list of known glycosyltransferases in lower eukaryotes, their activities and their respective genetic sequence. These genes are likely to be selected from the group of mannosyltransferases, e.g. 1,3 mannosyltransferases (e.g. MNN1 in S. cerevisiae) (Graham, 1991), 1,2 mannosyltransferases (e.g. KTR/KRE family from S. cerevisiae), 1,6 mannosyltransferases (OCH1 from S. cerevisiae), mannosylphosphate transferases and their regulators (MNN4 and MNN6 from S. cerevisiae) and additional enzymes that are involved in aberrant, i.e., non-human, glycosylation reactions. Many of these genes have in fact been deleted individually giving rise to viable phenotypes with altered glycosylation profiles. Examples are shown in Table 1.
  • Preferred lower eukaryotic host cells of the invention, as described herein to exemplify the required manipulation steps, are hypermannosylation-minus (och1) mutants of Pichia pastoris or K. lactis. Like other lower eukaryotes, P. pastoris processes Man9GlcNAc2 structures in the ER with an α-1,2-mannosidase to yield Man8GlcNAc2 (FIG. 1A). Through the action of several mannosyltransferases, this structure is then converted to hypermannosylated structures (Man>9GlcNAc2), also known as mannans (FIG. 35A). In addition, it has been found that P. pastoris is able to add non-terminal phosphate groups, through the action of mannosylphosphate transferases, to the carbohydrate structure. This differs from the reactions performed in mammalian cells, which involve the removal rather than addition of mannose sugars (FIG. 35A). It is of particular importance to eliminate the ability of the eukaryotic host cell, e.g., fungus, to hypermannosylate an existing Man8GlcNAc2 structure. This can be achieved by either selecting for a host cell that does not hypermannosylate or by genetically engineering such a cell.
  • Genes that are involved in the hypermannosylation process have been identified, e.g., in Pichia pastoris, and by creating mutations in these genes, one can reduce the production of “undesirable” glycoforms. Such genes can be identified by homology to existing mannosyltransferases or their regulators (e.g., OCH1, MNN4, MNN6, MNN1) found in other lower eukaryotes such as C. albicans, Pichia angusta or S. cerevisiae or by mutagenizing the host strain and selecting for a glycosylation phenotype with reduced mannosylation. Based on homologies amongst known mannosyltransferases and mannosylphosphate transferases, one may either design PCR primers (examples of which are shown in Table 2), or use genes or gene fragments encoding such enzymes as probes to identify homologs in DNA libraries of the target or a related organism. Alternatively, one may identify a functional homolog having mannosyltransferase activity by its ability to complement particular glycosylation phenotypes in related organisms.
    TABLE 2
    PCR Primers
    Target Gene(s)
    PCR primer A PCR primer B in P. pastoris Homologs
    ATGGCGAAGGCA TTAGTCCTTCCAA
    1,6- OCH1 S. cerevisiae,
    GATGGCAGT CTTCCTTC mannosyl- Pichia albicans
    (SEQ ID NO:3) (SEQ ID NO:4) transferase
    TAYTGGMGNGTN GCRTCNCCCCANC
    1,2 KTR/KRE family,
    GARCYNGAYATH KYTCRTA mannosyl- S. cerevisiae
    AA (SEQ ID NO:6) transferases
    (SEQ ID NO:5)

    Legend:

    M = A or C,

    R = A or G,

    W = A or T,

    S = C or G,

    Y = C or T,

    K = G or T,

    V = A or C or G,

    H = A or C or T,

    D = A or G or T,

    B = C or G or T,

    N = G or A or T or C.
  • To obtain the gene or genes encoding 1,6-mannosyltransferase activity in P. pastoris, for example, one would carry out the following steps: OCH1 mutants of S. cerevisiae are temperature sensitive and are slow growers at elevated temperatures. One can thus identify functional homologs of OCH1 in P. pastoris by complementing an OCH1 mutant of S. cerevisiae with a P. pastoris DNA or cDNA library. Mutants of S. cerevisiae are available, e.g., from Stanford University, and are commercially available from ResGen, Invitrogen Corp. (Carlsbad, Calif.). Mutants that display a normal growth phenotype at elevated temperature, after having been transformed with a P. pastoris DNA library, are likely to carry an OCH1 homolog of P. pastoris. Such a library can be created by partially digesting chromosomal DNA of P. pastoris with a suitable restriction enzyme and, after inactivating the restriction enzyme, ligating the digested DNA into a suitable vector, which has been digested with a compatible restriction enzyme.
  • Suitable vectors include, e.g., pRS314, a low copy (CEN6/ARS4) plasmid based on pBluescript containing the Trp1 marker (Sikorski and Hieter (1989) Genetics 122:19-27) and pFL44S, a high copy (2μ) plasmid based on a modified pUC 19 containing the URA3 marker (Bonneaud et al. (1991) Yeast 7:609-615). Such vectors are commonly used by academic researchers and similar vectors are available from a number of different vendors (e.g., Invitrogen (Carlsbad, Calif.); Pharmacia (Piscataway, N.J.); New England Biolabs (Beverly, Mass.)). Further examples include pYES/GS, 2μ origin of replication based yeast expression plasmid from Invitrogen, or Yep24 cloning vehicle from New England Biolabs.
  • After ligation of the chromosomal DNA and the vector, one may transform the DNA library into a strain of S. cerevisiae with a specific mutation and select for the correction of the corresponding phenotype. After sub-cloning and sequencing the DNA fragment that is able to restore the wild-type phenotype, one may use this fragment to eliminate the activity of the gene product encoded by OCH1 in P. pastoris using in vivo mutagenesis and/or recombination techniques well-known to those skilled in the art.
  • Alternatively, if the entire genomic sequence of a particular host cell, e.g., fungus, of interest is known, one may identify such genes simply by searching publicly available DNA databases, which are available from several sources, such as NCBI, Swissprot. For example, by searching a given genomic sequence or database with sequences from a known 1,6 mannosyltransferase gene (e.g., OCH1 from S. cerevisiae), one can identify genes of high homology in such a host cell genome which may (but do not necessarily) encode proteins that have 1,6-mannosyltransferase activity. Nucleic acid sequence homology alone is not enough to prove, however, that one has identified and isolated a homolog encoding an enzyme having the same activity. To date, for example, no data exist to show that an OCH1 deletion in P. pastoris eliminates the crucial initiating 1,6-mannosyltransferase activity (Martinet et al. (1998) Biotech. Letters 20(12):1171-1177; Contreras et al. WO 02/00856 A2). Thus, no data prove that the P. pastoris OCH1 gene homolog actually encodes that function. That demonstration is provided for the first time herein.
  • Homologs to several S. cerevisiae mannosyltransferases have been identified in P. pastoris using these approaches. Homologous genes often have similar functions to genes involved in the mannosylation of proteins in S. cerevisiae and thus their deletion may be used to manipulate the glycosylation pattern in P. pastoris or, by analogy, in any other host cell, e.g., fungus, plant, insect or animal cells, with similar glycosylation pathways.
  • The creation of gene knock-outs, once a given target gene sequence has been determined, is a well-established technique in the art and can be carried out by one of ordinary skill in the art (see, e.g., Rothstein (1991) Methods in Enzymology 194:281). The choice of a host organism may be influenced by the availability of good transformation and gene disruption techniques.
  • If several mannosyltransferases are to be knocked out, the method developed by Alani and Kleckner (1987) Genetics 116:541-545, for example, enables the repeated use of a selectable marker, e.g., the URA3 marker in yeast, to sequentially eliminate all undesirable endogenous mannosyltransferase activity. This technique has been refined by others but basically involves the use of two repeated DNA sequences, flanking a counter selectable marker. For example: URA3 may be used as a marker to ensure the selection of a transformants that have integrated a construct. By flanking the URA3 marker with direct repeats one may first select for transformants that have integrated the construct and have thus disrupted the target gene. After isolation of the transformants, and their characterization, one may counter select in a second round for those that are resistant to 5-fluoroorotic acid (5-FOA). Colonies that are able to survive on plates containing 5-FOA have lost the URA3 marker again through a crossover event involving the repeats mentioned earlier. This approach thus allows for the repeated use of the same marker and facilitates the disruption of multiple genes without requiring additional markers. Similar techniques for sequential elimination of genes adapted for use in another eukaryotic host cells with other selectable and counter-selectable markers may also be used.
  • Eliminating specific mannosyltransferases, such as 1,6 mannosyltransferase (OCH1) or mannosylphosphate transferases (MNN6, or genes complementing lbd mutants) or regulators (MNN4) in P. pastoris enables one to create engineered strains of this organism which synthesize primarily Man8GlcNAc2 and which can be used to further modify the glycosylation pattern to resemble more complex glycoform structures, e.g., those produced in mammalian, e.g., human cells. A preferred embodiment of this method utilizes DNA sequences encoding biochemical glycosylation activities to eliminate similar or identical biochemical functions in P. pastoris to modify the glycosylation structure of glycoproteins produced in the genetically altered P. pastoris strain.
  • Methods used to engineer the glycosylation pathway in yeasts as exemplified herein can be used in filamentous fungi to produce a preferred substrate for subsequent modification. Strategies for modifying glycosylation pathways in A. niger and other filamentous fungi, for example, can be developed using protocols analogous to those described herein for engineering strains to produce human-like glycoproteins in yeast. Undesired gene activities involved in 1,2 mannosyltransferase activity, e.g., KTR/KRE homologs, are modified or eliminated. A filamentous fungus, such as Aspergillus, is a preferred host because it lacks the 1,6 mannosyltransferase activity and as such, one would not expect a hypermannosylating gene activity, e.g. OCH1, in this host. By contrast, other desired activities (e.g., α-1,2-mannosidase, UDP-GlcNAc transporter, glycosyltransferase (GnT), galactosyltransferase (GalT) and sialyltransferase (ST)) involved in glycosylation are introduced into the host using the targeting methods of the invention.
  • Engineering or Selecting Hosts Having Diminished Initiating α-1,6 Mannosyltransferase Activity
  • In a preferred embodiment, the method of the invention involves making or using a host cell which is diminished or depleted in the activity of an initiating α-1,6-mannosyltransferase, i.e., an initiation specific enzyme that initiates outer chain mannosylation on the α-1,3 arm of the Man3GlcNAc2 core structure. In S. cerevisiae, this enzyme is encoded by the OCH1 gene. Disruption of the OCH1 gene in S. cerevisiae results in a phenotype in which N-linked sugars completely lack the poly-mannose outer chain. Previous approaches for obtaining mammalian-type glycosylation in fungal strains have required inactivation of OCH1 (see, e.g., Chiba et al. (1998) J. Biol. Chem. 273:26298-304). Disruption of the initiating α-1,6-mannosyltransferase activity in a host cell of the invention may be optional, however (depending on the selected host cell), as the Och1p enzyme requires an intact Man8GlcNAc2 for efficient mannose outer chain initiation. Thus, host cells selected or produced according to this invention which accumulate oligosaccharides having seven or fewer mannose residues may produce hypoglycosylated N-glycans that will likely be poor substrates for Och1p (see, e.g., Nakayama et al. (1 997) FEBS Lett. 412(3):547-50).
  • The OCH1 gene was cloned from P. pastoris (Example 1) and K. lactis (Example 9), as described. The nucleic acid and amino acid sequences of the OCH1 gene from K. lactis are set forth in SEQ ID NOs:7 and 8. Using gene-specific primers, a construct was made from each clone to delete the OCH1 gene from the genome of P. pastoris and K. lactis (Examples 1 and 9, respectively). Host cells depleted in initiating α-1,6-mannosyltransferase activity and engineered to produce N-glycans having a Man5GlcNAc2 carbohydrate structure were thereby obtained (see, e.g., FIGS. 5, 6, and 12; Examples 4 and 9).
  • Thus, in another embodiment, the invention provides an isolated nucleic acid molecule having a nucleic acid sequence comprising or consisting of at least forty-five, preferably at least 50, more preferably at least 60 and most preferably 75 or more nucleotide residues of the K. lactis OCH1 gene (SEQ ID NO: 7), and homologs, variants and derivatives thereof. The invention also provides nucleic acid molecules that hybridize under stringent conditions to the above-described nucleic acid molecules. Similarly, isolated polypeptides (including muteins, allelic variants, fragments, derivatives, and analogs) encoded by the nucleic acid molecules of the invention are provided. Also provided are vectors, including expression vectors, which comprise the above nucleic acid molecules of the invention, as described further herein. Similarly, host cells transformed with the nucleic acid molecules or vectors of the invention are provided.
  • The invention further provides methods of making or using a non-human eukaryotic host cell diminished or depleted in an alg gene activity (i.e., alg activities, including equivalent enzymatic activities in non-fungal host cells) and introducing into the host cell at least one glycosidase activity. In a preferred embodiment, the glycosidase activity is introduced by causing expression of one or more mannosidase activities within the host cell, for example, by activation of a mannosidase activity, or by expression from a nucleic acid molecule of a mannosidase activity, in the host cell.
  • In another embodiment, the method involves making or using a host cell diminished or depleted in the activity of one or more enzymes that transfer a sugar residue to the 1,6 arm of lipid-linked oligosaccharide precursors (FIG. 13). A host cell of the invention is selected for or is engineered by introducing a mutation in one or more of the genes encoding an enzyme that transfers a sugar residue (e.g., mannosylates) the 1,6 arm of a lipid-linked oligosaccharide precursor. The sugar residue is more preferably mannose, is preferably a glucose, GlcNAc, galactose, sialic acid, fucose or GlcNAc phosphate residue. In a preferred embodiment, the activity of one or more enzymes that mannosylate the 1,6 arm of lipid-linked oligosaccharide precursors is diminished or depleted. The method may further comprise the step of introducing into the host cell at least one glycosidase activity (see below).
  • In yet another embodiment, the invention provides a method for producing a human-like glycoprotein in a non-human host, wherein the glycoprotein comprises an N-glycan having at least two GlcNAcs attached to a trimannose core structure.
  • In each above embodiment, the method is directed to making a host cell in which the lipid-linked oligosaccharide precursors are enriched in ManXGlcNAc2 structures, where X is 3, 4 or 5 (FIG. 14). These structures are transferred in the ER of the host cell onto nascent polypeptide chains by an oligosaccharyl-transferase and may then be processed by treatment with glycosidases (e.g., α-mannosidases) and glycosyltransferases (e.g., GnTI) to produce N-glycans having GlcNAcManXGlcNAc2 core structures, wherein X is 3, 4 or 5, and is preferably 3 (FIGS. 14 and 15). As shown in FIG. 14, N-glycans having a GlcNAcManXGlcNAc2 core structure where X is greater than 3 may be converted to GlcNAcMan3GlcNAc2, e.g., by treatment with an α-1,3 and/or α-1,2-1,3 mannosidase activity, where applicable.
  • Additional processing of GlcNAcMan3GlcNAc2 by treatment with glycosyltransferases (e.g., GnTII) produces GlcNAc2Man3GlcNAc2 core structures which may then be modified, as desired, e.g., by ex vivo treatment or by heterologous expression in the host cell of a set of glycosylation enzymes, including glycosyltransferases, sugar transporters and mannosidases (see below), to become human-like N-glycans. Preferred human-like glycoproteins which may be produced according to the invention include those which comprise N-glycans having seven or fewer, or three or fewer, mannose residues; comprise one or more sugars selected from the group consisting of galactose, GlcNAc, sialic acid, and fucose; and comprise at least one oligosaccharide branch comprising the structure NeuNAc-Gal-GlcNAc-Man.
  • In one embodiment, the host cell has diminished or depleted Do1-P-Man:Man5GlcNAc2-PP-Do1 Mannosyltransferase activity, which is an activity involved in the first mannosylation step from Man5GlcNAc2-PP-Do1 to Man6GlcNAc2-PP-Do1 at the luminal side of the ER (e.g., ALG3 FIG. 13; FIG. 14). In S. cerevisiae, this enzyme is encoded by the ALG3 gene. As described above, S. cerevisiae cells harboring a leaky alg3-1 mutation accumulate Man5GlcNAc2-PP-Do1 and cells having a deletion in alg3 appear to transfer Man5GlcNAc2 structures onto nascent polypeptide chains within the ER. Accordingly, in this embodiment, host cells will accumulate N-glycans enriched in Man5GlcNAc2 structures which can then be converted to GlcNAc2Man3GlcNAc2 by treatment with glycosidases (e.g., with α-1,2 mannosidase, α-1,3 mannosidase, or α-1,2-1,3 mannosidase activities) and glycosyltransferase activites (e.g., GnTI, GnTII) (FIG. 14; FIG. 35B).
  • As described in Example 10, degenerate primers were designed based on an alignment of Alg3 protein sequences from S. cerevisiae, D. melanogaster and humans (H. sapiens) (FIGS. 16 and 17), and were used to amplify a product from P. pastoris genomic DNA. The resulting PCR product was used as a probe to identify and isolate a P. pastoris genomic clone comprising an open reading frame (ORF) that encodes a protein having 35% overall sequence identity and 53% sequence similarity to the S. cerevisiae ALG3 gene (FIGS. 18 and 19). This P. pastoris gene is referred to herein as “PpALG3”. The ALG3 gene was similarly identified and isolated from K. lactis (Example 10; FIGS. 20 and 21).
  • Thus, in another embodiment, the invention provides an isolated nucleic acid molecule having a nucleic acid sequence comprising or consisting of at least forty-five, preferably at least 50, more preferably at least 60 and most preferably 75 or more nucleotide residues of the P. pastoris ALG3 gene (FIG. 18) and the K. lactis ALG3 gene (FIG. 20), and homologs, variants and derivatives thereof. The invention also provides nucleic acid molecules that hybridize under stringent conditions to the above-described nucleic acid molecules. Similarly, isolated polypeptides (including muteins, allelic variants, fragments, derivatives, and analogs) encoded by the nucleic acid molecules of the invention are provided (P. pastoris and K. lactis ALG3 gene products are shown in FIGS. 18 and 20). In addition, also provided are vectors, including expression vectors, which comprise a nucleic acid molecule of the invention, as described further herein.
  • Using gene-specific primers, a construct was made to delete the PpALG3 gene from the genome of P. pastoris (Example 10). This strain was used to generate a host cell depleted in Do1-P-Man:Man5GlcNAc2-PP-Do1 Mannosyltransferase activity and produce lipid-linked Man5GlcNAc2-PP-Do1 precursors which are transferred onto nascent polypeptide chains to produce N-glycans having a Man5GlcNAc2 carbohydrate structure.
  • As described in Example 11, such a host cell may be engineered by expression of appropriate mannosidases to produce N-glycans having the desired Man3GlcNAc2 core carbohydrate structure. Expression of GnTs in the host cell (e.g., by targeting a nucleic acid molecule or a library of nucleic acid molecules as described below) enables the modified host cell to produce N-glycans having one or two GlcNAc structures attached to each arm of the Man3 core structure (i.e., GlcNAc1Man3GlcNAc2, GlcNAc2Man3GlcNAc2, or GlcNAc3Man3GlcNAc2; see FIG. 15). These structures may be processed further using the methods of the invention to produce human-like N-glycans on proteins which enter the secretion pathway of the host cell.
  • In a preferred embodiment, the method of the invention involves making or using a host cell which is both (a) diminished or depleted in the activity of an alg gene or in one or more activities that mannosylate N-glycans on the α-1,6 arm of the Man3GlcNAc2 (“Man3”) core carbohydrate structure; and (b) diminished or depleted in the activity of an initiating α-1,6-mannosyltransferase, i.e., an initiation specific enzyme that initiates outer chain mannosylation (on the α-1,3 arm of the Man3 core structure). In S. cerevisiae, this enzyme is encoded by the OCH1 gene. Disruption of the och1 gene in S. cerevisiae results in a phenotype in which N-linked sugars completely lack the poly-mannose outer chain. Previous approaches for obtaining mammalian-type glycosylation in fungal strains have required inactivation of OCH1 (see, e.g., Chiba et al. (1998) J. Biol. Chem. 273:26298-304). Disruption of the initiating α-1,6-mannosyltransferase activity in a host cell of the invention is optional, however (depending on the selected host cell), as the Och1p enzyme requires an intact Man8GlcNAc for efficient mannose outer chain initiation. Thus, the host cells selected or produced according to this invention, which accumulate lipid-linked oligosaccharides having seven or fewer mannose residues will, after transfer, produce hypoglycosylated N-glycans that will likely be poor substrates for Och1p (see, e.g., Nakayama et al. (1997) FEBS Lett. 412(3):547-50).
  • Engineering or Selecting Hosts Having N-Acetylglucosaminyltransferase III Activity
  • The invention additionally provides a method for producing a human-like glycoprotein in a lower eukaryotic host cell by expressing an N-acetylglucosaminyltransferase III activity (including a full-length enzyme, homologs, variants, derivatives, and catalytically active fragments thereof). In one embodiment, a host cell (e.g., P. pastoris) is engineered to produce more human-like N-glycans, e.g., by activation of an N-acetylglucosaminyltransferase III activity or by expression from a nucleic acid molecule of an N-acetylglucosaminyltransferase III activity. Using well-known techniques in the art, gene-specific primers are designed to complement the homologous regions of a GnTIII gene, preferably a mammalian GnTIII gene (e.g., mouse GnTIII) (FIG. 24), sequences for which are readily available in the art (e.g., Genbank Accession No. L39373) and are PCR amplified.
  • In one embodiment, the invention provides a method for producing a human-like glycoprotein in a lower eukaryote (e.g., P. pastoris), wherein the glycoprotein comprises an N-glycan exhibiting a bisecting GlcNAc on a trimannose or trimannosyl (Man3GlcNAc2) core structure. In this embodiment, GlcNAcMan3GlcNAc2 (which may be produced by reacting a trimannose core with N-acetylglucosaminyltransferase I (“GnTI”) activity, but which is typically produced by trimming of GlcNAcMan5GlcNAc2 by an α-1,3/α-1,6-mannosidase activity, such as Mannosidase II (Hamilton et al. (2003) Science 301:1244-46)) is reacted with an N-acetylglucosaminyltransferase III activity to produce a bisected GlcNAc2Man3GlcNAc2. Accordingly, the invention provides GnTIII activity, which transfers β-1,4 GlcNAc onto substrates that are capable of accepting the bisecting GlcNAc in lower eukaryotes.
  • In another embodiment, the invention provides a method for producing a human-like glycoprotein in a lower eukaryote (e.g., P. pastoris), wherein the glycoprotein comprises an N-glycan exhibiting a bisecting GlcNAc on a trimannose or trimannosyl (Man3GlcNAc2) core structure having at least two GlcNAcs attached to the trimannose core. In this embodiment, Man3GlcNAc2 is reacted with a GnTI activity and then with an N-acetylglucosaminyltransferase II (“GnTII”) activity and a GnTIII activity (in either order) to produce a bisected GlcNAc3Man3GlcNAc2 (FIG. 38). It should be appreciated that the bisected trimannosyl core structure of this embodiment may also contain an additional mannosyl group in place of a GlcNAc residue. For example, GlcNAcMan4GlcNAc2 may be reacted with a GnTIII activity to produce a bisected GlcNAc2Man4GlcNAc2.
  • The invention also provides a method for producing a more human-like glycoprotein in a lower eukaryote (e.g. P. pastoris), wherein the glycoprotein produced comprises an N-glycan having at least two GlcNAcs attached to a pentamannose core structure (Man5GlcNAc2) and which exhibits a bisected N-glycan. Accordingly, in this embodiment, a pentamannose core structure (Man5GlcNAc2) is reacted with GnTIII activity to produce a bisected GlcNAcMan5GlcNAc2 and GlcNAc2Man5GlcNAc2 structure.
  • In an alternative embodiment, a pentamannose core structure produced via the mutation of och1 and alg3 genes is reacted with α1,2-mannosidase, GnTI, GnTII and GnTIII activities and UDP-GlcNAc to produce a bisected GlcNAc3Man3GlcNAc2 glycan (FIG. 35B). In another embodiment, a pentamannose core structure is reacted with GnTI and GnTIII activities (in either order or in combination) to produce a bisected GlcNAc2Man5GlcNAc2 structure (FIG. 37).
  • In a more preferred embodiment, using the combinatorial DNA library method of the invention, as described below, a pVA53 construct comprising the S. cerevisiae MNN2(s) leader (GenBank Accession No. NP009571) fused to a catalytically active GnTIII domain from mouse (GnTIII Δ32) is expressed in a P. pastoris strain YSH-1 (Example 13) thereby producing N-glycans having a bisected GlcNAc2Man5GlcNAc2 structure (Example 20). FIG. 26 (bottom) displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in the above-mentioned strain, which is designated PBP26 (FIG. 36), exhibiting a predominant peak at 1666 m/z [a], which corresponds to bisected GlcNAc2Man5GlcNAc2. (For comparison, FIG. 26 (top) displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in strain YSH-1 lacking the pVA53 construct. The predominant peak at 1461 m/z [d] corresponds to the unmodified glycan: GlcNAcMan5GlcNAc2.) Accordingly, in one embodiment, a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 50 mole % of a GlcNAc2Man5GlcNAc2 or at least 50 mole % of a GlcNAc2Man3GlcNAc2 structure having a bisecting GlcNAc. The mole percent of the glycans is in reference to percent of total neutral glycans as detected by MALDI-TOF. It is understood that if, for example, GlcNAc2Man3GlcNAc2 having a bisecting GlcNAc is produced at 20% and GlcNAc3Man3GlcNAc2 is produced at 25% on a target protein, the total amount of transiently produced GlcNAc2Man3GlcNAc2 having a bisecting GlcNAc is 45%, because GlcNAc3Man3GlcNAc2 is a product of a GlcNAc2Man3GlcNAc2 having a bisecting GlcNAc further reacted with GnTII.
  • Similarly, in another embodiment, a pVA55 construct comprising the S. cerevisiae MNN2(1) leader (GenBank Accession No. NP009571) fused to a catalytically active GnTIII domain from mouse (GnTIII A32) is expressed in a P. pastoris strain (YSH-1) thereby producing N-glycans GlcNAcMan5GlcNAc2 and bisected N-glycans GlcNAc2Man5GlcNAc2 structure. As shown in FIG. 27 (bottom), these structures correspond to peaks at 1463 m/z and 1667 m/z, respectively. (For comparison, FIG. 27 (top) displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in strain YSH-1 lacking the pVA53 construct. The predominant peak corresponds to unmodified GlcNAcMan5GlcNAc2 at 1461 m/z [d].) Accordingly, in another embodiment, a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 20 mole % of a GlcNAc2Man5GlcNAc2 or at least 20 mole % of a GlcNAc2Man3GlcNAc2 structure having a bisecting GlcNAc.
  • In an even more preferred embodiment, a pVA53 construct comprising the S. cerevisiae MNN2(s) leader (GenBank Accession No. NP009571) fused to a catalytically active GnTIII domain from mouse (GnTIII Δ32) is expressed in a P. pastoris strain YSH-44 (Example 15) thereby producing N-glycans having a bisected GlcNAc3Man3GlcNAc2 structure (Example 20). FIG. 30 displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in the above-mentioned strain designated as YSH-57, exhibiting a predominant peak at 1542 m/z [y], which corresponds to the bisected glycan GlcNAc3Man3GlcNAc2. (For comparison, FIG. 29 displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in strain YSH-44 lacking the pVA53 construct. The predominant peak at 1356 m/z [x] in FIG. 29 corresponds to the unmodified glycan: GlcNAc2Man3GlcNAc2.) Accordingly, in one embodiment, a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 80 mole % of a GlcNAc3Man3GlcNAc2 structure having a bisecting GlcNAc. The mole percent of the glycans is in reference to percent of total neutral glycans as detected by MALDI-TOF.
  • Alternatively, in another embodiment, a pVA53 construct comprising the S. cerevisiae MNN2(s) leader (GenBank Accession No. NP009571) fused to a catalytically active GnTIII domain from mouse (GnTIII Δ32) is expressed in a P. pastoris strain (PBP6-5) (Example 11) thereby producing N-glycans having a GlcNAc2Man3GlcNAc2 and a bisected GlcNAc3Man3GlcNAc2 structure. As shown in FIG. 32, these structures correspond to peaks at 1340 m/z and 1543 m/z, respectively. Accordingly, in another embodiment, a host of the present invention is characterized by its ability to produce, at least transiently, N-glycans which exhibit at least 20 mole % of a GlcNAc3Man3GlcNAc2 structure having a bisecting GlcNAc in an alg3 mutant host cell.
  • The invention provides methods for producing a human-like glycoprotein in a lower eukaryote, wherein the glycoprotein comprises a Man5GlcNAc2 core structure or a Man3GlcNAc2 core structure, and wherein the core structure is further modified by two or more GlcNAcs. In some embodiments of the invention, 10% or more of the core structures are modified by the two or more GlcNAcs. In other preferred embodiments, 20%, 30%, 40%, 50%, 60%, 70%, 80% or even more of the core structures are so modified. In a highly preferred embodiment, one of the GlcNAcs is a bisecting GlcNAc.
  • In another aspect of the invention, a combinatorial nucleic acid library which encodes at least one GnTIII catalytic domain is used to express a GnTIII activity in a lower eukaryotic host cell (Example 18). Preferably, a library of the invention comprises a sublibrary of leader sequences fused in frame to a single nucleic acid molecule or a sublibrary of nucleic acid molecules comprising GnTIII sequences, one or more of which encode a catalytic domain having GnTIII activity in the host cell. Alternatively, a single nucleic acid molecule or a sublibrary of nucleic acid molecules comprising leader sequences is fused in frame to a sublibrary of nucleic acid molecules comprising GnTIII sequences, one or more of which encode a catalytic domain having GnTIII activity in the host cell. (See below.) Expression of these and other such combinatorial libraries is performed in a host cell which expresses a target glycoprotein whose N-glycan structures are analyzed to determine whether and how much GnTIII is expressed. A wide range of catalytically active GnTIII enzymes may be produced in a host cell using the methods and libraries of the invention. It is this aspect of the invention that allows a skilled artisan to create and delinate between GnTIII enzymes having little or no activity and those enzymes that are actively expressed and which produce predominant levels of a desired bisected oligosaccharide intermediate such as GlcNAc2Man5GlcNAc2, GlcNAc3Man3GlcNAc2 or GlcNAc2Man3GlcNAc2 in the host cells.
  • As described further below, the proper targeting of an enzyme responsible for a given step in the glycosylation pathway to the appropriate subcellular location and the sufficiency of the enzyme's activity at the particular pH of that subcellular location are important factors in the production of glycoproteins having N-glycans with the desired structures. The use of combinatorial libraries of fusion proteins to generate diverse populations of enzyme chimeras and the screening of these libraries in transformed cells provides a powerful method to identify host strains with the activity of interest in the appropriate location. In preferred embodiments of the invention, the enzyme activity is located such that an N-glycan-containing glycoprotein expressed in the cell is capable of reacting with the activity during the secretion process.
  • Not all combinations of leader/catalytic domains produce desired enzyme activities however. A wide variety of leader/catalytic domain combinations is created, only a few of which may be useful in producing the presently desired intermediates. The present invention, nevertheless, encompasses even those combinations that do not presently exhibit a desired enzymatic activity in the exemplified host cell. FIG. 28 (bottom) shows a pVB51 construct comprising the K. lactis GNT(s) leader (GenBank Accession No. AF106080) fused to a catalytically active GnTIII domain from mouse (GnTIII A32) expressed in a P. pastoris strain YSH-1, which does not readily exhibit GnTIII activity. (For comparison, FIG. 28 (top) displays the MALDI-TOF spectrum of N-glycans released from a kringle 3 protein expressed in strain YSH-1 lacking the pVA53 construct. The predominant peak corresponds to unmodified GlcNAcMan5GlcNAc2 at 1461 m/z.) The predominant peak in FIG. 28 (bottom) at 1463 m/z, which correlates to the mass of GlcNAcMan5GlcNAc2, is observed. A second peak at 1726 m/z, which does not correlate to the mass of GlcNAc2Man5GlcNAc2 is also observed. It is contemplated that these and other such combinations may be useful, with or without slight modifications using techniques well known in the art, when they are expressed, e.g., in other host cells including those which have been modified to produce human-like glycoforms.
  • The use of combinatorial libraries to generate diverse populations of enzyme chimeras and the screening of these libraries in transformed cells further allows strains to be identified in which the enzyme activity is substantially intracellular. Example 6, below, provides an example of assay conditions useful for measuring extracellular α-1,2-mannosidase activity. Examples 22 and 23 also provide examples of assays for glycosyltransferase activity (GnTIII) in the medium. See also Table 9, below, and Choi et al. (20033 Proc. Natl. Acad. Sci. U.S.A. 100(9):5022-27. For purposes of the invention, an enzyme activity is substantially intracellular when less than 10% of the enzyme activity is measurable in the extracellular medium.
  • As described in Examples 11, 12, 13, 14, 15, and 19-21, a host cell may be engineered by the expression of appropriate glycosyltransferases (e.g., N-acetylglucosaminyltransferase) to produce N-glycans having the desired carbohydrate structures (e.g., GlcNAc2Man3GlcNAc2, GlcNAc3Man3GlcNAc2). Expression of GnTs in the host cell (e.g., by targeting a nucleic acid molecule or a library of nucleic acid molecules as described below and in Choi et al. (2003) Proc. Natl. Acad. Sci. US.A. 100(9):5022-27 and WO 02/00879) enables the modified host cell to produce N-glycans having the bisecting GlcNAc on the middle mannose. These structures may be processed further using the methods of the invention to produce human-like N-glycans on proteins which enter the secretion pathway of the host cell.
  • In a more preferred embodiment, co-expression of appropriate UDP-sugar-transporter(s) and transferase(s) will cap the terminal α-1,6 and α-1,3 residues as well as the middle mannose with GlcNAc, resulting in the precursor for mammalian-type complex (e.g. GlcNAc3Man3GlcNAc2) and hybrid N-glycosylation. These peptide-bound N-linked oligosaccharide chains then serve as a precursor for further modification to a mammalian-type oligosaccharide structure. Subsequent expression of galactosyl-tranferases and genetically engineering the capacity to transfer sialylic acid to the termini (see FIG. 1B) will produce a mammalian-type (e.g., human-like) N-glycan structure.
  • Host Cells of the Invention
  • A preferred host cell of the invention is a lower eukaryotic cell, e.g., yeast, a unicellular and multicellular or filamentous fungus. However, a wide variety of host cells are envisioned as being useful in the methods of the invention. Plant cells or insect cells, for instance, may be engineered to express a human-like glycoprotein according to the invention. Likewise, a variety of non-human, mammalian host cells may be altered to express more human-like or otherwise altered glycoproteins using the methods of the invention. As one of skill in the art will appreciate, any eukaryotic host cell (including a human cell) may be used in conjunction with a library of the invention to express one or more chimeric proteins which is targeted to a subcellular location, e.g., organelle, in the host cell where the activity of the protein is modified, and preferably is enhanced. Such a protein is preferably—but need not necessarily be—an enzyme involved in protein glycosylation, as exemplified herein. It is envisioned that any protein coding sequence may be targeted and selected for modified activity in a eukaryotic host cell using the methods described herein.
  • Lower eukaryotes that are able to produce glycoproteins having the attached N-glycan Man5GlcNAc2 are particularly useful because (a) lacking a high degree of mannosylation (e.g., greater than 8 mannoses per N-glycan, or especially 30-40 mannoses), they show reduced immunogenicity in humans; and (b) the N-glycan is a substrate for further glycosylation reactions to form an even more human-like glycoform, e.g., by the action of GlcNAc transferase I (FIG. 1B; β1,2 GnTI) to form GlcNAcMan5GlcNAc2. A yield is obtained of greater than 30 mole %, more preferably a yield of 50, 60, 70, 80, 90, or even 100 mole %, glycoproteins with N-glycans having a Man5GlcNAc2 structure. In a preferred embodiment, more than 50% of the Man5GlcNAc2 structure is shown to be a substrate for a GnTI activity and can serve as such a substrate in vivo.
  • Preferred lower eukaryotes of the invention include but are not limited to: Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reseei, Chrysosporium lucknowense, Fusarium sp. Fusarium gramineum, Fusarium venenatum, and Neurospora crassa.
  • In each above embodiment, the method is directed to making a host cell in which the oligosaccharide precursors are enriched in Man5GlcNAc2. These structures are desirable because they may then be processed by treatment in vitro, for example, using the method of Maras and Contreras, U.S. Pat. No. 5,834,251. In a preferred embodiment, however, precursors enriched in Man5GlcNAc2 are processed by at least one further glycosylation reaction in vivo—with glycosidases (e.g., α-mannosidases) and glycosyltransferases (e.g., GnTI)—to produce human-like N-glycans. Oligosaccharide precursors enriched in Man5GlcNAc2, for example, are preferably processed to those having GlcNAcManXGlcNAc2 core structures, wherein X is 3, 4 or 5, and is preferably 3. N-glycans having a GlcNAcManXGlcNAc2 core structure where X is greater than 3 may be converted to GlcNAcMan3GlcNAc2, e.g., by treatment with an α-1,3 and/or α-1,6 mannosidase activity, where applicable. Additional processing of GlcNAcMan3GlcNAc2 by treatment with glycosyltransferases (e.g., GnTII) produces GlcNAc2Man3GlcNAc2 core structures which may then be modified, as desired, e.g., by ex vivo treatment or by heterologous expression in the host cell of additional glycosylation enzymes, including glycosyltransferases, sugar transporters and mannosidases (see below), to become human-like N-glycans.
  • Preferred human-like glycoproteins which may be produced according to the invention include those which comprise N-glycans having seven or fewer, or three or fewer, mannose residues; and which comprise one or more sugars selected from the group consisting of galactose, GlcNAc, sialic acid, and fucose.
  • Another preferred non-human host cell of the invention is a lower eukaryotic cell, e.g., a unicellular or filamentous fungus, which is diminished or depleted in the activity of one or more alg gene activities (including an enzymatic activity which is a homolog or equivalent to an alg activity). Another preferred host cell of the invention is diminished or depleted in the activity of one or more enzymes (other than alg activities) that mannosylate the α-1,6 arm of a lipid-linked oligosaccharide structure.
  • While lower eukaryotic host cells are preferred, a wide variety of host cells having the aforementioned properties are envisioned as being useful in the methods of the invention. Plant cells, for instance, may be engineered to express a human-like glycoprotein according to the invention. Likewise, a variety of non-human, mammalian host cells may be altered to express more human-like glycoproteins using the methods of the invention. An appropriate host cell can be engineered, or one of the many such mutants already described in yeasts may be used. A preferred host cell of the invention, as exemplified herein, is a hypermannosylation-minus (OCH1) mutant in Pichia pastoris which has further been modified to delete the alg3 gene.
  • The invention additionally provides lower eukaryotic host cells capable of producing glycoproteins having bisected N-glycans, such as bisected GlcNAcMan5GlcNAc2, GlcNAc2Man5GlcNAc2, GlcNAc2Man3GlcNAc2, and, preferably, GlcNAc3Man3GlcNAc2. In a preferred embodiment of the invention, the host cells comprise a GnTIII activity. In a more preferred embodiment, the host cells further comprise one or more activities selected from: GnTI, GnTII, GnTIV, and GnTV. Preferred host cells express GnTI, GnTII, and GnTIII. Other preferred host cells additionally express GnTIV and/or GnTV. Even more preferably, the one or more GnT activities of the host cells are substantially intracellular.
  • Thus, in preferred embodiments of the invention, host cells comprising the one or more GnT activities produce N-glycans comprising structures, including but not limited to GlcNAcMan3GlcNAc2, GlcNAcMan4GlcNAc2, or GlcNAcMan5GlcNAc2, that are capable of reacting with a GnTIII enzyme activity to produce corresponding bisected N-glycans. The enzyme activities thereby convert glycoproteins containing these N-glycans into forms with new and more desirable properties. Because GnTIII is currently understood to inhibit additional GnT activity in mammalian cells, the skilled artisan should appreciate that sequential glycosylation reaction may or may not be of importance. The present invention contemplates, however, the addition of GnTI and GnTIII in either order or together. It should also be understood that other enzyme activities within the cell, such as, e.g., one or more desired mannosidase activities (e.g., α 1,2 mannosidase, Mannosidase I, Mannosidase II), may act in concert with the GnT activities to generate yet other human-like glycoproteins of interest (see FIG. 1B).
  • In a preferred embodiment, a mannosidase II or a catalytically active fragment thereof is introduced into the host cell to trim the α1,3 and α1,6 mannose containing arms of a bisected pentamannose core structure such as GlcNAc2Man5GlcNAc2. The resulting glycans (e.g., bisected GlcNAc2Man4GlcNAc2 and GlcNAc2Man3GlcNAc2) are preferred substrates for subsequent human-like N-glycan modification.
  • In another embodiment of the invention, the host cells comprise a Man5GlcNAc2 core structure or a Man3GlcNAc2 core structure modified by two or more GlcNAcs. It should be understood that either core structure may include further modifications in addition to the modification by GlcNAc. Preferably, 10% or more of the core structures are modified by GlcNAcs. Most preferably, 20%, 30%, 40%, 50%, 60%, 70%, 80% or even more of the core structures contain the GlcNAc modification.
  • Formation of Complex N-glycans
  • Formation of complex N-glycan synthesis is a sequential process by which specific sugar residues are removed and attached to the core oligosaccharide structure. In higher eukaryotes, this is achieved by having the substrate sequentially exposed to various processing enzymes. These enzymes carry out specific reactions depending on their particular location within the entire processing cascade. This “assembly line” consists of ER, early, medial and late Golgi, and the trans Golgi network all with their specific processing environment. To re-create the processing of human glycoproteins in the Golgi and ER of lower eukaryotes, numerous enzymes (e.g., glycosyltransferases, glycosidases, phosphatases and transporters) have to be expressed and specifically targeted to these organelles, and preferably, in a location so that they function most efficiently in relation to their environment as well as to other enzymes in the pathway.
  • Because one goal of the methods described herein is to achieve a robust protein production strain that is able to perform well in an industrial fermentation process, the integration of multiple genes into the host cell chromosome involves careful planning. As described above, one or more genes which encode enzymes known to be characteristic of non-human glycosylation reactions are preferably deleted. The engineered cell strain is transformed with a range of different genes encoding desired activities, and these genes are transformed in a stable fashion, thereby ensuring that the desired activity is maintained throughout the fermentation process.
  • Any combination of the following enzyme activities may be engineered singly or multiply into the host using methods of the invention: sialyltransferases, mannosidases, fucosyltransferases, galactosyltransferases, GlcNAc transferases, ER and Golgi specific transporters (e.g. syn- and antiport transporters for UDP-galactose and other precursors), other enzymes involved in the processing of oligosaccharides, and enzymes involved in the synthesis of activated oligosaccharide precursors such as UDP-galactose and CMP-N-acetylneuraminic acid. Preferably, enzyme activities are introduced on one or more nucleic acid molecules (see also below). Nucleic acid molecules may be introduced singly or multiply, e.g., in the context of a nucleic acid library such as a combinatorial library of the invention. It is to be understood, however, that single or multiple enzymatic activities may be introduced into a host cell in any fashion, including but not limited to protein delivery methods and/or by use of one or more nucleic acid molecules without necessarily using a nucleic acid library or combinatorial library of the invention.
  • Expression Of Glycosyltransferases To Produce Complex N-glycans:
  • With DNA sequence information, the skilled artisan can clone DNA molecules encoding GnT activities (e.g., Example 3, 8, 11, 15, and 18). Using standard techniques well-known to those of skill in the art, nucleic acid molecules encoding GnTI, II, III, IV or V (or encoding catalytically active fragments thereof) may be inserted into appropriate expression vectors under the transcriptional control of promoters and other expression control sequences capable of driving transcription in a selected host cell of the invention, e.g., a fungal host such as Pichia sp., Kluyveromyces sp. and Aspergillus sp., as described herein, such that one or more of these mammalian GnT enzymes may be actively expressed in a host cell of choice for production of a human-like complex glycoprotein (e.g., Examples 8, 20, and 21).
  • Several individual glycosyltransferases have been cloned and expressed in S. cerevisiae (GalT, GnTI), Aspergillus nidulans (GnTI) and other fungi, without however demonstrating the desired outcome of “humanization” on the glycosylation pattern of the organisms (Yoshida et al. (1999) Glycobiology 9(l):53-8; Kalsner et al. (1995) Glycoconj. J 12(3):360-370). It was speculated that the carbohydrate structure required to accept sugars by the action of such glycosyltransferases was not present in sufficient amounts, which most likely contributed to the lack of complex N-glycan formation.
  • A preferred method of the invention provides the functional expression of a GnT, such as GnTI, GnTII, and GnTIII, in the early, medial or late Golgi apparatus, as well as ensuring a sufficient supply of UDP-GlcNAc (e.g., by expression of a UDP-GlcNAc transporter; see Examples below).
  • Methods for Providing Sugar Nucleotide Precursors to the Golgi Apparatus:
  • For a glycosyltransferase to function satisfactorily in the Golgi, the enzyme requires a sufficient concentration of an appropriate nucleotide sugar, which is the high-energy donor of the sugar moiety added to a nascent glycoprotein. In humans, the full range of nucleotide sugar precursors (e.g., UDP-N-acetylglucosamine, UDP-N-acetylgalactosamine, CMP-N-acetylneuraminic acid, UDP-galactose, etc.) are generally synthesized in the cytosol and transported into the Golgi, where they are attached to the core oligosaccharide by glycosyltransferases.
  • To replicate this process in non-human host cells, such as lower eukaryotes, sugar nucleoside specific transporters have to be expressed in the Golgi to ensure adequate levels of nucleoside sugar precursors (Sommers and Hirschberg (1981) J. Cell Biol. 91(2):A406-A406; Sommers and Hirschberg (1982) J. Biol. Chem. 257(18):811-817; Perez and Hirschberg (1987) Methods in Enzymology 138:709-715). Nucleotide sugars may be provided to the appropriate compartments, e.g., by expressing in the host microorganism an exogenous gene encoding a sugar nucleotide transporter. The choice of transporter enzyme is influenced by the nature of the exogenous glycosyltransferase being used. For example, a GlcNAc transferase may require a UDP-GlcNAc transporter, a fucosyltransferase may require a GDP-fucose transporter, a galactosyltransferase may require a UDP-galactose transporter, and a sialyltransferase may require a CMP-sialic acid transporter.
  • The added transporter protein conveys a nucleotide sugar from the cytosol into the Golgi apparatus, where the nucleotide sugar may be reacted by the glycosyltransferase, e.g., to elongate an N-glycan. The reaction liberates a nucleoside diphosphate or monophosphate, e.g., UDP, GDP, or CMP. Nucleoside monophosphates can be directly exported from the Golgi in exchange for nucleoside triphosphate sugars by an antiport mechanism. Accumulation of a nucleoside diphosphate, however, inhibits the further activity of a glycosyltransferase. As this reaction appears to be important for efficient glycosylation, it is frequently desirable to provide an expressed copy of a gene encoding a nucleotide diphosphatase. The diphosphatase (specific for UDP or GDP as appropriate) hydrolyzes the diphosphonucleoside to yield a nucleoside monosphosphate and inorganic phosphate.
  • Suitable transporter enzymes, which are typically of mammalian origin, are described below. Such enzymes may be engineered into a selected host cell using the methods of the invention.
  • In another example, α2,3- or α2,6-sialyltransferase caps galactose residues with sialic acid in the trans-Golgi and TGN of humans leading to a mature form of the glycoprotein (FIG. 1B). To reengineer this processing step into a metabolically engineered yeast or fungus will require (1) α2,3- or α2,6-sialyltransferase activity and (2) a sufficient supply of CMP-N-acetyl neuraminic acid, in the late Golgi of yeast. To obtain sufficient a 2,3-sialyltransferase activity in the late Golgi, for example, the catalytic domain of a known sialyltransferase (e.g. from humans) has to be directed to the late Golgi in fungi (see above). Likewise, transporters have to be engineered to allow the transport of CMP-N-acetyl neuraminic acid into the late Golgi. There is currently no indication that fungi synthesize or can even transport sufficient amounts of CMP-N-acetyl neuraminic acid into the Golgi. Consequently, to ensure the adequate supply of substrate for the corresponding glycosyltransferases, one has to metabolically engineer the production of CMP-sialic acid into the fungus.
  • UDP-N-Acetylglucosamine
  • The cDNA of human UDP-N-acetylglucosamine transporter, which was recognized through a homology search in the expressed sequence tags database (dbEST), has been cloned (Ishida (1999) J. Biochem. 126(1):68-77). The mammalian Golgi membrane transporter for UDP-N-acetylglucosamine was cloned by phenotypic correction with cDNA from canine kidney cells (MDCK) of a recently characterized Kluyveromyces lactis mutant deficient in Golgi transport of the above nucleotide sugar (Guillen et al. (1998) Proc. Natl. Acad. Sci. USA 95(14):7888-7892). Results demonstrate that the mammalian Golgi UDP-GlcNAc transporter gene has all of the necessary information for the protein to be expressed and targeted functionally to the Golgi apparatus of yeast and that two proteins with very different amino acid sequences may transport the same solute within the same Golgi membrane (Guillen et al. (1998) Proc. Natl. Acad. Sci. USA 95(14):7888-7892).
  • Accordingly, one may incorporate the expression of a UDP-GlcNAc transporter in a host cell by means of a nucleic acid construct which may contain, for example: (1) a region by which the transformed construct is maintained in the cell (e.g., origin of replication or a region that mediates chromosomal integration), (2) a marker gene that allows for the selection of cells that have been transformed, including counterselectable and recyclable markers such as ura3 or T-urf13 (Soderholm et al. (2001) Biotechniques 31(2):306-10) or other well characterized selection-markers (e.g., his4, bla, Sh ble etc.), (3) a gene or fragment thereof encoding a functional UDP-GlcNAc transporter (e.g., from K. lactis, (Abeijon, (1996) Proc. Natl. Acad. Sci. USA. 93:5963-5968), or from H. sapiens (Ishida et al. (1996) J. Biochem. (Tokyo) 120(6):1074-8), and (4) a promoter activating the expression of the above mentioned localization/catalytic domain fusion construct library.
  • GDP-Fucose
  • The rat liver Golgi membrane GDP-fucose transporter has been identified and purified by Puglielli and Hirschberg (1999) J. Biol. Chem. 274(50):35596-35600. The corresponding gene has not been identified, however, N-terminal sequencing can be used for the design of oligonucleotide probes specific for the corresponding gene. These oligonucleotides can be used as probes to clone the gene encoding for GDP-fucose transporter.
  • UDP-Galactose
  • Two heterologous genes, gma12(+) encoding alpha 1,2-galactosyltransferase ( alpha 1,2 GalT) from Schizosaccharomyces pombe and (hUGT2) encoding human UDP-galactose (UDP-Gal) transporter, have been functionally expressed in S. cerevisiae to examine the intracellular conditions required for galactosylation. Correlation between protein galactosylation and UDP-galactose transport activity indicated that an exogenous supply of UDP-Gal transporter, rather than alpha 1,2 GaiT played a key role for efficient galactosylation in S. cerevisiae (Kainuma (1999) Glycobiology 9(2):133-141). Likewise, an UDP-galactose transporter from S. pombe was cloned (Segawa (1999) FEBS Letters 451 (3):295-298).
  • CMP-N-Acetylneuraminic Acid (CMP-Sialic Acid)
  • Human CMP-sialic acid transporter (hCST) has been cloned and expressed in Lec 8 CHO cells (Aoki et al. (1999) J. Biochem. (Tokyo) 126(5):940-50; Eckhardt et al. (1997) Eur. J Biochem. 248(1):187-92). The functional expression of the murine CMP-sialic acid transporter was achieved in Saccharomyces cerevisiae (Berninsone et al. (1997) J. Biol. Chem. 272(19):12616-9). Sialic acid has been found in some fungi, however it is not clear whether the chosen host system will be able to supply sufficient levels of CMP-Sialic acid. Sialic acid can be either supplied in the medium or alternatively fungal pathways involved in sialic acid synthesis can also be integrated into the host genome.
  • Expression of Diphosphatases:
  • When sugars are transferred onto a glycoprotein, either a nucleoside diphosphate or monophosphate is released from the sugar nucleotide precursors. While monophosphates can be directly exported in exchange for nucleoside triphosphate sugars by an antiport mechanism, diphosphonucleosides (e.g. GDP) have to be cleaved by phosphatases (e.g. GDPase) to yield nucleoside monophosphates and inorganic phosphate prior to being exported. This reaction appears to be important for efficient glycosylation, as GDPase from S. cerevisiae has been found to be necessary for mannosylation. However, the enzyme only has 10% of the activity towards UDP (Berninsone et al. (1994) J. Biol. Chem. 269(l):207-21 1). Lower eukaryotes often do not have UDP-specific diphosphatase activity in the Golgi as they do not utilize UDP-sugar precursors for glycoprotein synthesis in the Golgi. Schizosaccharomyces pombe, a yeast which adds galactose residues to cell wall polysaccharides (from UDP-galactose), was found to have specific UDPase activity, further suggesting the requirement for such an enzyme (Berninsone et al. (1994) J. Biol. Chem. 269(1):207-211). UDP is known to be a potent inhibitor of glycosyltransferases and the removal of this glycosylation side product is important to prevent glycosyltransferase inhibition in the lumen of the Golgi (Khatara et al. (1974) Eur. J. Biochem. 44:537-560).
  • Methods For Altering N-Glycans in a Host By Expressing A Targeted Enzymatic Activity From a Nucleic Acid Molecule
  • The present invention further provides a method for producing a human-like glycoprotein in a non-human host cell comprising the step of introducing into the cell one or more nucleic acid molecules which encode an enzyme or enzymes for production of the Man5GlcNAc2 carbohydrate structure. In one preferred embodiment, a nucleic acid molecule encoding one or more mannosidase activities involved in the production of Man5GlcNAc2 from Man8GlcNAc2 or Man9GlcNAc2 is introduced into the host. The invention additionally relates to methods for making altered glycoproteins in a host cell comprising the step of introducing into the host cell a nucleic acid molecule which encodes one or more glycosylation enzymes or activities. Preferred enzyme activities are selected from the group consisting of UDP-GlcNAc transferase, UDP-galactosyltransferase, GDP-fucosyltransferase, CMP-sialyltransferase, UDP-GlcNAc transporter, UDP-galactose transporter, GDP-fucose transporter, CMP-sialic acid transporter, and nucleotide diphosphatases. In a particularly preferred embodiment, the host is selected or engineered to express two or more enzymatic activities in which the product of one activity increases substrate levels of another activity, e.g., a glycosyltransferase and a corresponding sugar transporter, e.g., GnTI and UDP-GlcNAc transporter activities. In another preferred embodiment, the host is selected or engineered to expresses an activity to remove products which may inhibit subsequent glycosylation reactions, e.g. a UDP- or GDP-specific diphosphatase activity.
  • Preferred methods of the invention involve expressing one or more enzymatic activities from a nucleic acid molecule in a host cell and comprise the step of targeting at least one enzymatic activity to a desired subcellular location (e.g., an organelle) by forming a fusion protein comprising a catalytic domain of the enzyme and a cellular targeting signal peptide, e.g., a heterologous signal peptide which is not normally ligated to or associated with the catalytic domain. The fusion protein is encoded by at least one genetic construct (“fusion construct”) comprising a nucleic acid fragment encoding a cellular targeting signal peptide ligated in the same translational reading frame (“in-frame”) to a nucleic acid fragment encoding an enzyme (e.g., glycosylation enzyme), or catalytically active fragment thereof.
  • The targeting signal peptide component of the fusion construct or protein is preferably derived from a member of the group consisting of: membrane-bound proteins of the ER or Golgi, retrieval signals, Type II membrane proteins, Type I membrane proteins, membrane spanning nucleotide sugar transporters, mannosidases, sialyltransferases, glucosidases, mannosyltransferases and phosphomannosyltransferases.
  • The catalytic domain component of the fusion construct or protein is preferably derived from a glycosidase, mannosidase or a glycosyltransferase activity derived from a member of the group consisting of GnTI, GnTII, GnTIII, GnTIV, GnTV, GnTVI, GalT, Fucosyltransferase and Sialyltransferase. The catalytic domain preferably has a pH optimum within 1.4 pH units of the average pH optimum of other representative enzymes in the organelle in which the enzyme is localized, or has optimal activity at a pH between 5.1 and 8.0. In a preferred embodiment, the catalytic domain encodes a mannosidase selected from the group consisting of C. elegans mannosidase IA, C. elegans mannosidase IB, D. melanogaster mannosidase IA, H. sapiens mannosidase IB, P. citrinum mannosidase I, mouse mannosidase IA, mouse mannosidase IB, A. nidulans mannosidase IA, A. nidulans mannosidase IB, A. nidulans mannosidase IC, mouse mannosidase II, C. elegans mannosidase II, H. sapiens mannosidase II, mannosidase Iix, and mannosidase III.
  • Selecting a Glycosylation Enzyme: pH Optima and Subcellular Localization
  • In one embodiment of the invention, a human-like glycoprotein is made efficiently in a non-human eukaryotic host cell by introducing into a subcellular compartment of the cell a glycosylation enzyme selected to have a pH optimum similar to the pH optima of other enzymes in the targeted subcellular compartment. For example, most enzymes that are active in the ER and Golgi apparatus of S. cerevisiae have pH optima that are between about 6.5 and 7.5 (see Table 3). Because the glycosylation of proteins is a highly evolved and efficient process, the internal pH of the ER and the Golgi is likely also in the range of about 6-8. All previous approaches to reduce mannosylation by the action of recombinant mannosidases in fungal hosts, however, have introduced enzymes that have a pH optimum of around pH 5.0 (Martinet et al. (1998) Biotech. Letters 20(12): 1171-1177, and Chiba et al. (1998) J. Biol. Chem. 273(41): 26298-26304). At pH 7.0, the in vitro determined activity of those mannosidases is reduced to less than 10%, which is likely insufficient activity at their point of use, namely, the ER and early Golgi, for the efficient in vivo production of Man5GlcNAc2 on N-glycans.
  • Accordingly, a preferred embodiment of this invention targets a selected glycosylation enzyme (or catalytic domain thereof), e.g., an α-mannosidase, to a subcellular location in the host cell (e.g., an organelle) where the pH optimum of the enzyme or domain is within 1.4 pH units of the average pH optimum of other representative marker enzymes localized in the same organelle(s). The pH optimum of the enzyme to be targeted to a specific organelle should be matched with the pH optimum of other enzymes found in the same organelle to maximize the activity per unit enzyme obtained. Table 3 summarizes the activity of mannosidases from various sources and their respective pH optima. Table 4 summarizes their typical subcellular locations.
    TABLE 3
    Mannosidases and their pH optimum.
    pH
    Source Enzyme optimum Reference
    Aspergillus saitoi α-1,2-mannosidase 5.0 Ichishima et al. (1999)
    Biochem. J. 339(Pt
    3): 589-597
    Trichoderma reesei α-1,2-mannosidase 5.0 Maras et al. (2000) J.
    Biotechnol. 77(2-3): 255-263
    Penicillium α-D-1,2- 5.0 Yoshida et al. (1993)
    citrinum mannosidase Biochem. J. 290(Pt
    2): 349-354
    C. elegans α-1,2-mannosidase 5.5 see FIG. 11
    Aspergillus α-1,2-mannosidase 6.0 Eades and Hintz (2000)
    nidulans Gene 255(1): 25-34
    Homo sapiens α-1,2-mannosidase 6.0
    IA(Golgi)
    Homo sapiens IB α-1,2-mannosidase 6.0
    (Golgi)
    Lepidopteran Type I α-1,2-Man6- 6.0 Ren et al. (1995)
    insect cells mannosidase Biochem. 34(8): 2489-2495
    Homo sapiens α-D-mannosidase 6.0 Chandrasekaran et al.
    (1984) Cancer Res.
    44(9): 4059-68
    Xanthomonas α-1,2,3-mannosidase 6.0 U.S. Pat. No. 6,300,113
    manihotis
    Mouse IB (Golgi) α-1,2-mannosidase 6.5 Schneikert and
    Herscovics (1994)
    Glycobiology. 4(4): 445-50
    Bacillus sp. α-D-1,2- 7.0 Maruyama et al. (1994)
    (secreted) mannosidase Carbohydrate Res.
    251: 89-98
  • In a preferred embodiment, a particular enzyme or catalytic domain is targeted to a subcellular location in the host cell by means of a chimeric fusion construct encoding a protein comprising a cellular targeting signal peptide not normally associated with the enzymatic domain. Preferably, an enzyme or domain is targeted to the ER, the early, medial or late Golgi, or the trans Golgi apparatus of the host cell.
  • In a more preferred embodiment, the targeted glycosylation enzyme is a mannosidase, glycosyltransferase or a glycosidase. In an especially preferred embodiment, mannosidase activity is targeted to the ER or cis Golgi, where the early reactions of glycosylation occur. While this method is useful for producing a human-like glycoprotein in a non-human host cell, it will be appreciated that the method is also useful more generally for modifying carbohydrate profiles of a glycoprotein in any eukaryotic host cell, including human host cells.
  • Targeting sequences which mediate retention of proteins in certain organelles of the host cell secretory pathway are well-known and described in the scientific literature and public databases, as discussed in more detail below with respect to libraries for selection of targeting sequences and targeted enzymes. Such subcellular targeting sequences may be used alone or in combination to target a selected glycosylation enzyme (or catalytic domain thereof) to a particular subcellular location in a host cell, i.e., especially to one where the enzyme will have enhanced or optimal activity based on pH optima or the presence of other stimulatory factors.
  • When one attempts to trim high mannose structures to yield Man5GlcNAc2 in the ER or the Golgi apparatus of a host cell such as S. cerevisiae, for example, one may choose any enzyme or combination of enzymes that (1) has a sufficiently close pH optimum (i.e., between pH 5.2 and pH 7.8), and (2) is known to generate, alone or in concert, the specific isomeric Man5GlcNAc2 structure required to accept subsequent addition of GlcNAc by GnTI. Any enzyme or combination of enzymes that is shown to generate a structure that can be converted to GlcNAcMan5GlcNAc2 by GnTI in vitro would constitute an appropriate choice. This knowledge may be obtained from the scientific literature or experimentally. For example, one may determine whether a potential mannosidase can convert Man8GlcNAc2-2AB (2-aminobenzamide) to Man5GlcNAc2-AB and then verify that the obtained Man5GlcNAc2-2AB structure can serve a substrate for GnTI and UDP-GlcNAc to give GlcNAcMan5GlcNAc2 in vitro. Mannosidase IA from a human or murine source, for example, would be an appropriate choice (see, e.g., Example 4). Examples described herein utilize 2-aminobenzamide labeled N-linked oligomannose followed by HPLC analysis to make this determination.
    TABLE 4
    Cellular location and pH optima of various glycosylation-related
    enzymes of S. cerevisiae.
    Lo- pH op-
    Gene Activity cation timum Reference(s)
    KTR1 α-1,2 Golgi 7.0 Romero et al.
    mannosyltransferase (1997) Biochem.
    J. 321(Pt 2): 289-295
    MNS1 α-1,2-mannosidase ER 6.5 Lipari et al.
    (1994)
    Glycobiology.
    Oct; 4(5): 697-702
    CWH41 glucosidase I ER 6.8
    mannosyltransferase Golgi 7-8 Lehele and
    Tanner (1974)
    Biochim.
    Biophys. Acta
    350(1): 225-235
    KRE2 α-1,2 Golgi 6.5-9.0 Romero et al.
    mannosyltransferase (1997) Biochem.
    J. 321(Pt 2): 289-295
  • Accordingly, a glycosylation enzyme such as an α-1,2-mannosidase enzyme used according to the invention has an optimal activity at a pH of between 5.1 and 8.0. In a preferred embodiment, the enzyme has an optimal activity at a pH of between 5.5 and 7.5. The C. elegans mannosidase enzyme, for example, works well in the methods of the invention and has an apparent pH optimum of about 5.5). Preferred mannosidases include those listed in Table 3 having appropriate pH optima, e.g. Aspergillus nidulans, Homo sapiens IA (Golgi), Homo sapiens IB (Golgi), Lepidopteran insect cells (IPLB-SF21 AE), Homo sapiens, mouse IB (Golgi), Xanthomonas manihotis, Drosophila melanogaster and C. elegans.
  • An experiment which illustrates the pH optimum for an α-1,2-mannosidase enzyme is described in Example 7. A chimeric fusion protein BB27-2 (Saccharomyces MNN10 (s)/C. elegans mannosidase IB Δ31), which leaks into the medium was subjected to various pH ranges to determine the optimal activity of the enzyme. The results of the experiment show that the α-1,2-mannosidase has an optimal pH of about 5.5 for its function (FIG. 11).
  • In a preferred embodiment, a single cloned mannosidase gene is expressed in the hostlorganism. However, in some cases it may be desirable to express several different mannosidase genes, or several copies of one particular gene, in order to achieve adequate production of Man5GlcNAc2. In cases where multiple genes are used, the encoded mannosidases preferably all have pH optima within the preferred range of about 5.1 to about 8.0, or especially between about 5.5 and about 7.5. Preferred mannosidase activities include α-1,2-mannosidases derived from mouse, human, Lepidoptera, Aspergillus nidulans, or Bacillus sp., C. elegans, D. melanogaster, P. citrinum, X laevis or A. nidulans.
  • In Vivo Alteration of Host Cell Glycosylation Using a Combinatorial DNA Library
  • Certain methods of the invention are preferably (but need not necessarily be) carried out using one or more nucleic acid libraries. An exemplary feature of a combinatorial nucleic acid library of the invention is that it comprises sequences encoding cellular targeting signal peptides and sequences encoding proteins to be targeted (e.g., enzymes or catalytic domains thereof, including but not limited to those which mediate glycosylation).
  • In one embodiment, a combinatorial nucleic acid library comprises: (a) at least two nucleic acid sequences encoding different cellular targeting signal peptides; and (b) at least one nucleic acid sequence encoding a polypeptide to be targeted. In another embodiment, a combinatorial nucleic acid library comprises: (a) at least one nucleic acid sequence encoding a cellular targeting signal peptide; and (b) at least two nucleic acid sequences encoding a polypeptide to be targeted into a host cell. As described further below, a nucleic acid sequence derived from (a) and a nucleic acid sequence derived from (b) are ligated to produce one or more fusion constructs encoding a cellular targeting signal peptide functionally linked to a polypeptide domain of interest. One example of a functional linkage is when the cellular targeting signal peptide is ligated to the polypeptide domain of interest in the same translational reading frame (“in-frame”).
  • In a preferred embodiment, a combinatorial DNA library expresses one or more fusion proteins comprising cellular targeting signal peptides ligated in-frame to catalytic enzyme domains. The encoded fusion protein preferably comprises a catalytic domain of an enzyme involved in mammalian- or human-like modification of N-glycans. In a more preferred embodiment, the catalytic domain is derived from an enzyme selected from the group consisting of mannosidases, glycosyltransferases and other glycosidases which is ligated in-frame to one or more targeting signal peptides. The enzyme domain may be exogenous and/or endogenous to the host cell. A particularly preferred signal peptide is one normally associated with a protein that undergoes ER to Golgi transport.
  • The combinatorial DNA library of the present invention may be used for producing and localizing in vivo enzymes involved in mammalian- or human-like N-glycan modification. The fusion constructs of the combinatorial DNA library are engineered so that the encoded enzymes are localized in the ER, Golgi or the trans-Golgi network of the host cell where they are involved in producing particular N-glycans on a glycoprotein of interest. Localization of N-glycan modifying enzymes of the present invention is achieved through an anchoring mechanism or through protein-protein interaction where the localization peptide constructed from the combinatorial DNA library localizes to a desired organelle of the secretory pathway such as the ER, Golgi or the trans Golgi network.
  • An example of a useful N-glycan, which is produced efficiently and in sufficient quantities for further modification by human-like (complex) glycosylation reactions is Man5GlcNAc2. A sufficient amount of Man5GlcNAc2 is needed on a glycoprotein of interest for further human-like processing in vivo (e.g., more than 30 mole %). The Man5GlcNAc2 intermediate may be used as a substrate for further N-glycan modification to produce GlcNAcMan5GlcNAc2 (FIG. 1B; see above). Accordingly, the combinatorial DNA library of the present invention may be used to produce enzymes that subsequently produce GlcNAcMan5GlcNAc2, or other desired complex N-glycans, in a useful quantity.
  • A further aspect of the fusion constructs produced using the combinatorial DNA library of the present invention is that they enable sufficient and often near complete intracellular N-glycan trimming activity in the engineered host cell. Preferred fusion constructs produced by the combinatorial DNA library of the invention encode a glycosylation enzyme, e.g., a mannosidase, which is effectively localized to an intracellular host cell compartment and thereby exhibits very little and preferably no extracellular activity. The preferred fusion constructs of the present invention that encode a mannosidase enzyme are shown to localize where the N-glycans are modified, namely, the ER and the Golgi. The fusion enzymes of the present invention are targeted to such particular organelles in the secretory pathway where they localize and act upon N-glycans such as Man8GlcNAc2 to produce Man5GlcNAc2 on a glycoprotein of interest.
  • GnTIII fusion constructs generated from a combinatorial DNA library to produce bisected glycans were assayed to determine any extracellular activity: An example of a GnTIII fusion constructs exhibiting in vivo alteration of host cell glycosylation is designated pVA53. After transforming P. pastoris YSH-1 with the fusion construct pVA53, the supernatant was tested to detect any ex vivo GnTIII activity. FIG. 33 shows no apparent change in the standard substrate GlcNAcMan5GlcNAc2 under conditions that would reveal extracellular GnTIII activity in the medium (Example 22). Similarly, FIG. 34 shows no detectable extracellular GnTIII activity in the medium in P. pastoris YSH-57 reacting with the substrate GlcNAc2Man3GlcNAc2 (Example 23).
  • Enzymes produced by the combinatorial DNA library of the present invention can modify N-glycans on a glycoprotein of interest as shown for K3 or IFN-β proteins expressed in P. pastoris, as shown in FIGS. 5, 6, and 25-34 (see also Examples 2, 4, and 18-23). It is, however, appreciated that other types of glycoproteins, without limitation, including erythropoietin, cytokines such as interferon-α, interferon-β, interferon-γ, interferon-ω, and granulocyte-CSF, coagulation factors such as factor VIII, factor IX, and human protein C, soluble IgE receptor α-chain, IgG, IgG fragments, IgM, urokinase, chymase, urea trypsin inhibitor, IGF-binding protein, epidermal growth factor, growth hormone-releasing factor, annexin V fusion protein, angiostatin, vascular endothelial growth factor-2, myeloid progenitor inhibitory factor-1, osteoprotegerin, α-1 antitrypsin, DNase II, and α-feto proteins may be glycosylated in this way.
  • Constructing a Combinatorial DNA Library of Fusion Constructs:
  • A combinatorial DNA library of fusion constructs features one or more cellular targeting signal peptides (“targeting peptides”) generally derived from N-terminal domains of native proteins (e.g., by making C-terminal deletions). Some targeting peptides, however, are derived from the C-terminus of native proteins (e.g. SEC12). Membrane-bound proteins of the ER or the Golgi are preferably used as a source for targeting peptide sequences. These proteins have sequences encoding a cytosolic tail (ct), a transmembrane domain (tmd) and a stem region (sr) which are varied in length. These regions are recognizable by protein sequence alignments and comparisons with known homologs and/or other localized proteins (e.g., comparing hydrophobicity plots).
  • The targeting peptides are indicated herein as short (s), medium (m) and long (1) relative to the parts of a type II membrane. The targeting peptide sequence indicated as short (s) corresponds to the transmembrane domain (tmd) of the membrane-bound protein. The targeting peptide sequence indicated as long (1) corresponds to the length of the transmembrane domain (tmd) and the stem region (sr). The targeting peptide sequence indicated as medium (m) corresponds to the transmembrane domain (tmd) and approximately half the length of the stem region (sr). The catalytic domain regions are indicated herein by the number of nucleotide deletion with respect to its wild-type glycosylation enzyme.
  • Sub-Libraries
  • In some cases a combinatorial nucleic acid library of the invention may be assembled directly from existing or wild-type genes. In a preferred embodiment, the DNA library is assembled from the. fusion of two or more sub-libraries. By the in-frame ligation of the sub-libraries, it is possible to create a large number of novel genetic constructs encoding useful targeted protein domains such as those which have glycosylation activities.
  • Catalytic Domain Sub-Libraries Encoding Glycosylation Activities
  • One useful sub-library includes DNA sequences encoding enzymes such as glycosidases (e.g., mannosidases), glycosyltransferases (e.g., fucosyl-transferases, galactosyltransferases, glucosyltransferases), GlcNAc transferases and sialyltransferases. Catalytic domains may be selected from the host to be engineered, as well as from other related or unrelated organisms. Mammalian, plant, insect, reptile, algal or fungal enzymes are all useful and should be chosen to represent a broad spectrum of biochemical properties with respect to temperature and pH optima. In a preferred embodiment, genes are truncated to give fragments some of which encode the catalytic domains of the enzymes. By removing endogenous targeting sequences, the enzymes may then be redirected and expressed in other cellular loci.
  • The choice of such catalytic domains may be guided by the knowledge of the particular environment in which the catalytic domain is subsequently to be active. For example, if a particular glycosylation enzyme is to be active in the late Golgi, and all known enzymes of the host organism in the late Golgi have a certain pH optimum, or the late Golgi is known to have a particular pH, then a catalytic domain is chosen which exhibits adequate, and preferably maximum, activity at that pH, as discussed above.
  • Targeting Peptide Sequence Sub-Libraries
  • Another useful sub-library includes nucleic acid sequences encoding targeting signal peptides that result in localization of a protein to a particular location within the ER, Golgi, or trans Golgi network. These targeting peptides may be selected from the host organism to be engineered as well as from other related or unrelated organisms. Generally such sequences fall into three categories: (1) N-terminal sequences encoding a cytosolic tail (ct), a transmembrane domain (tmd) and part or all of a stem region (sr), which together or individually anchor proteins to the inner (lumenal) membrane of the Golgi; (2) retrieval signals which are generally found at the C-terminus such as the HDEL (SEQ ID NO:41) or KDEL (SEQ ID NO:42) tetrapeptide; and (3) membrane spanning regions from various proteins, e.g., nucleotide sugar transporters, which are known to localize in the Golgi.
  • In the first case, where the targeting peptide consists of various elements (ct, tmd and sr), the library is designed such that the ct, the tmd and various parts of the stem region are represented. Accordingly, a preferred embodiment of the sub-library of targeting peptide sequences includes ct, tmd, and/or sr sequences from membrane-bound proteins of the ER or Golgi. In some cases it may be desirable to provide the sub-library with varying lengths of sr sequence. This may be accomplished by PCR using primers that bind to the 5′ end of the DNA encoding the cytosolic region and employing a series of opposing primers that bind to various parts of the stem region.
  • Still other useful sources of targeting peptide sequences include retrieval signal peptides, e.g. the tetrapeptides HDEL or KDEL, which are typically found at the C-terminus of proteins that are transported retrograde into the ER or Golgi. Still other sources of targeting peptide sequences include (a) type II membrane proteins, (b) the enzymes listed in Table 3, (c) membrane spanning nucleotide sugar transporters that are localized in the Golgi, and (d) sequences referenced in Table 5.
    TABLE 5
    Sources of useful compartmental targeting sequences
    Gene or Location of
    Sequence Organism Function Gene Product
    MNSI A. nidulans α-1,2-mannosidase ER
    MNSI A. niger α-1,2-mannosidase ER
    MNSI S. cerevisiae α-1,2-mannosidase ER
    GLSI S. cerevisiae glucosidase ER
    GLSI A. niger glucosidase ER
    GLSI A. nidulans glucosidase ER
    HDEL Universal in retrieval signal ER
    at C- fungi
    terminus
    SEC12 S. cerevisiae COPII vesicle protein ER/Golgi
    SEC12 A. niger COPII vesicle protein ER/Golgi
    OCH1 S. cerevisiae 1,6-mannosyltransferase Golgi (cis)
    OCH1 P. pastoris 1,6-mannosyltransferase Golgi (cis)
    MNN9 S. cerevisiae 1,6-mannosyltransferase Golgi
    complex
    MNN9 A. niger undetermined Golgi
    VAN1 S. cerevisiae undetermined Golgi
    VAN1 A. niger undetermined Golgi
    ANP1 S. cerevisiae undetermined Golgi
    HOCI S. cerevisiae undetermined Golgi
    MNN10 S. cerevisiae undetermined Golgi
    MNN10 A. niger undetermined Golgi
    MNN11 S. cerevisiae undetermined Golgi (cis)
    MNN11 A. niger undetermined Golgi (cis)
    MNT1 S. cerevisiae 1,2-mannosyltransferase Golgi (cis,
    medial)
    KTR1 P. pastoris undetermined Golgi (medial)
    KRE2 P. pastoris undetermined Golgi (medial)
    KTR3 P. pastoris undetermined Golgi (medial)
    MNN2 S. cerevisiae 1,2-mannosyltransferase Golgi (medial)
    KTR1 S. cerevisiae undetermined Golgi (medial)
    KTR2 S. cerevisiae undetermined Golgi (medial)
    MNN1 S. cerevisiae 1,3-mannosyltransferase Golgi (trans)
    MNN6 S. cerevisiae Phosphomannosyltransferase Golgi (trans)
    2,6 ST H. sapiens 2,6-sialyltransferase trans Golgi
    network
    UDP- S. pombe UDP-Gal transporter Golgi
    Gal T
  • In any case, it is highly preferred that targeting peptide sequences are selected which are appropriate for the particular enzymatic activity or activities to function optimally within the sequence of desired glycosylation reactions. For example, in developing a modified microorganism capable of terminal sialylation of nascent N-glycans, a process which occurs in the late Golgi in humans, it is desirable to utilize a sub-library of targeting peptide sequences derived from late Golgi proteins. Similarly, the trimming of Man8GlcNAc2by an α-1,2-mannosidase to give Man5GlcNAc2 is an early step in complex N-glycan formation in humans (FIG. 1B). It is therefore desirable to have this reaction occur in the ER or early Golgi of an engineered host microorganism. A sub-library encoding ER and early Golgi retention signals is used.
  • A series of fusion protein constructs (i.e., a combinatorial DNA library) is then constructed by functionally linking one or a series of targeting peptide sequences to one or a series of sequences encoding catalytic domains. In a preferred embodiment, this is accomplished by the in-frame ligation of a sub-library comprising DNA encoding targeting peptide sequences (above) with a sub-library comprising DNA encoding glycosylation enzymes or catalytically active fragments thereof (see below).
  • The resulting library comprises synthetic genes encoding targeting peptide sequence-containing fusion proteins. In some cases it is desirable to provide a targeting peptide sequence at the N-terminus of a fusion protein, or in other cases at the C-terminus. In some cases, targeting peptide sequences may be inserted within the open reading frame of an enzyme, provided the protein structure of individual folded domains is not disrupted. Each type of fusion protein is constructed (in a step-wise directed or semi-random fashion) and optimal constructs may be selected upon transformation of host cells and characterization of glycosylation patterns in transformed cells using methods of the invention.
  • Alteration of Host Cell Glycosylation Using Fusion Constructs From Combinatorial Libraries:
  • The construction of a preferred combinatorial DNA library is illustrated schematically in FIG. 2 and described in Example 4. The fusion construct may be operably linked to a multitude of vectors, such as expression vectors well-known in the art. A wide variety of such fusion constructs were assembled using representative activities as shown in Table 6. Combinations of targeting peptide/catalytic domains may be assembled for use in targeting mannosidase, glycosyltransferase and glycosidase activities in the ER, Golgi, and the trans Golgi network according to the invention. Surprisingly, the same catalytic domain may have no effect to a very profound effect on N-glycosylation patterns, depending on the type of targeting peptide used (see, e.g., Table 7, Example 4).
  • Mannosidase Fusion Constructs
  • A representative example of a mannosidase fusion construct derived from a combinatorial DNA library of the invention is pFB8, which a truncated Saccharomyces SEC12(m) targeting peptide (988-1296 nucleotides of SEC12 from SwissProt P11655) ligated in-frame to a 187 N-terminal amino acid deletion of a mouse α-mannosidase IA (Genbank AN 6678787). The nomenclature used herein, thus, refers to the targeting peptide/catalytic domain region of a glycosylation enzyme as Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187. The encoded fusion protein localizes in the ER by means of the SEC12 targeting peptide sequence while retaining its mannosidase catalytic domain activity and is capable of producing in vivo N-glycans having a Man5GlcNAc2 structure (Example 4; FIGS. 6F and 7B).
  • The fusion construct pGC5, Saccharomyces MNS1 (m)/mouse mannosidase IB Δ99, is another example of a fusion construct having intracellular mannosidase trimming activity (Example 4; FIGS. 5D and 8B). Fusion construct pBC18-5 (Saccharomyces VAN1 (s)/C. elegans mannosidase IB Δ80) is yet another example of an efficient fusion construct capable of producing N-glycans having a Man5GlcNAc2 structure in vivo. By creating a combinatorial DNA library of these and other such mannosidase fusion constructs according to the invention, a skilled artisan may distinguish and select those constructs having optimal intracellular trimming activity from those having relatively low or no activity. Methods using combinatorial DNA libraries of the invention are advantageous because only a select few mannosidase fusion constructs may produce a particularly desired N-glycan in vivo.
  • In addition, mannosidase trimming activity may be specific to a particular protein of interest. Thus, it is to be further understood that not all targeting peptide/mannosidase catalytic domain fusion constructs may function equally well to produce the proper glycosylation on a glycoprotein of interest. Accordingly, a protein of interest may be introduced into a host cell transfected with a combinatorial DNA library to identify one or more fusion constructs which express a mannosidase activity optimal for the protein of interest. One skilled in the art will be able to produce and select optimal fusion construct(s) using the combinatorial DNA library approach described herein.
  • It is apparent, moreover, that other such fusion constructs exhibiting localized active mannosidase catalytic domains (or more generally, domains of any enzyme) may be made using techniques such as those exemplified in Example 4 and described herein. It will be a matter of routine experimentation for one skilled in the art to make and use the combinatorial DNA library of the present invention to optimize, for example, Man5GlcNAc2 production from a library of fusion constructs in a particular expression vector introduced into a particular host cell.
  • Glycosyltransferase Fusion Constructs
  • Similarly, a glycosyltransferase combinatorial DNA library was made using the methods of the invention. A combinatorial DNA library of sequences derived from glycosyltransferase I (GnTI) activities were assembled with targeting peptides and screened for efficient production in a lower eukaryotic host cell of a GlcNAcMan5GlcNAc2 N-glycan structure on a marker glycoprotein. A fusion construct shown to produce GlcNAcMan5GlcNAc2 (pPB1O4), Saccharomyces MNN9(s)/human GnTI A38 was identified (Example 8). A wide variety of such GnTI fusion constructs were assembled (Example 8, Table 10). Other combinations of targeting peptide/GnTI catalytic domains can readily be assembled by making a combinatorial DNA library. It is also apparent to one skilled in the art that other such fusion constructs exhibiting glycosyltransferase activity may be made as demonstrated in Example 8. It will be a matter of routine experimentation for one skilled in the art to use the combinatorial DNA library method described herein to optimize GlcNAcMan5GlcNAc2 production using a selected fusion construct in a particular expression vector and host cell line.
  • As stated above for mannosidase fusion constructs, not all targeting peptide/GnTI catalytic domain fusion constructs will function equally well to produce the proper glycosylation on a glycoprotein of interest as described herein. However, one skilled in the art will be able to produce and select optimal fusion construct(s) using a DNA library approach as described herein. Example 8 illustrates a preferred embodiment of a combinatorial DNA library comprising targeting peptides and GnTI catalytic domain fusion constructs involved in producing glycoproteins with predominantly GlcNAcMan5GlcNAc2 structure.
  • Using Multiple Fusion Constructs to Alter Host Cell Glycosylation
  • In another example of using the methods and libraries of the invention to alter host cell glycosylation, a P. pastoris strain with an OCH1 deletion that expresses a reporter protein (K3) was transformed with multiple fusion constructs isolated from combinatorial libraries of the invention to convert high mannose N-glycans to human-like N-glycans (Example 8). First, the mannosidase fusion construct pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187) was transformed into a P. pastoris strain lacking 1,6 initiating mannosyltransferases activity (i.e., och1 deletion; Example 1). Second, pPB103 comprising a K. lactis MNN2-2 gene (Genbank AN AF 106080) encoding an UDP-GlcNAc transporter was constructed to increase further production of GlcNAcMan5GlcNAc2. The addition of the UDP-GlcNAc transporter increased production of GlcNAcMan5GlcNAc2 significantly in the P. pastoris strain as illustrated in FIG. 10B. Third, pPB104 comprising Saccharomyces MNN9 (s)/human GnTI Δ38 was introduced into the strain. This P. pastoris strain is referred to as “PBP-3.” (See FIG. 36.)
  • It is understood by one skilled in the art that host cells such as the above-described yeast strains can be sequentially transformed and/or co-transformed with one or more expression vectors. It is also understood that the order of transformation is not particularly relevant in producing the glycoprotein of interest. The skilled artisan recognizes the routine modifications of the procedures disclosed herein may provide improved results in the production of the glycoprotein of interest.
  • The importance of using a particular targeting peptide sequence with a particular catalytic domain sequence becomes readily apparent from the experiments described herein. The combinatorial DNA library provides a tool for constructing enzyme fusions that are involved in modifying N-glycans on a glycoprotein of interest, which is especially useful in producing human-like glycoproteins. (Any enzyme fusion, however, may be selected using libraries and methods of the invention.) Desired transformants expressing appropriately targeted, active α-1,2-mannosidase produce K3 with N-glycans of the structure Man5GlcNAc2 as shown in FIGS. 5D and 5E. This confers a reduced molecular mass to the cleaved glycan compared to the K3 of the parent OCH1 deletion strain, as was detected by MALDI-TOF mass spectrometry in FIG. 5C.
  • Similarly, the same approach was used to produce another secreted glycoprotein: IFN-β comprising predominantly Man5GlcNAc2. The Man5GlcNAc2 was removed by PNGase digestion (Papac et al. (1998) Glycobiology 8:445-454) and subjected to MALDI-TOF as shown in FIGS. 6A-6F. A single prominent peak at 1254 (m/z) confirms Man5GlcNA2 production on IFN-β in FIGS. 6E (pGC5) (Saccharomyces MNS1 (m)/mouse mannosidase IB Δ99) and 6F (pFB8) (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187). Furthermore, in the P. pastoris strain PBP-3 comprising pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187), pPB104 (Saccharomyces MNN9 (s)/human GnTI Δ38) and pPB103 (K. lactis MNN2-2 gene), the hybrid N-glycan GlcNAcMan5GlcNAc2[b] was detected by MALDI-TOF (FIG. 10).
  • After identifying transformants with a high degree of mannose trimming, additional experiments were performed to confirm that mannosidase (trimming) activity occurred in vivo and was not predominantly the result of extracellular activity in the growth medium (Example 6; FIGS. 7-9).
  • Although the present invention is exemplified using a P. pastoris host organism, it is understood by those skilled in the art that other eukaryotic host cells, including other species of yeast and fungal hosts, may be altered as described herein to produce human-like glycoproteins. The techniques described herein for identification and disruption of undesirable host cell glycosylation genes, e.g. OCH1, is understood to be applicable for these and/or other homologous or functionally related genes in other eukaryotic host cells such as other yeast and fungal strains. As described in Example 9, och1 mnn1 genes were deleted from K. lactis to engineer a host cell leading to N-glycans that are completely converted to Man5GlcNAc2 by 1,2-mannosidase (FIG. 12C).
  • The MNN1 gene was cloned from K. lactis as described in Example 9. The nucleic acid and deduced amino acid sequences of the K. lactis MNN1 gene are shown in SEQ ID NOs:43 and 44, respectively. Using gene-specific primers, a construct was made to delete the MNN1 gene from the genome of K. lactis (Example 9). Host cells depleted in och1 and mnn1 activities produce N-glycans having a Man9GlcNAc2 carbohydrate structure (see, e.g., FIG. 12B). Such host cells may be engineered further using, e.g., methods and libraries of the invention, to produce mammalian- or human-like glycoproteins.
  • Thus, in another embodiment, the invention provides an isolated nucleic acid molecule having a nucleic acid sequence comprising or consisting of at least forty-five, preferably at least 50, more preferably at least 60 and most preferably 75 or more nucleotide residues of the K. lactis MNN1 gene (SEQ ID NO: 43), and homologs, variants and derivatives thereof. The invention also provides nucleic acid molecules that hybridize under stringent conditions to the above-described nucleic acid molecules. Similarly, isolated polypeptides (including muteins, allelic variants, fragments, derivatives, and analogs) encoded by the nucleic acid molecules of the invention are provided. In addition, also provided are vectors, including expression vectors, which comprise a nucleic acid molecule of the invention, as described further herein. Similarly host cells transformed with the nucleic acid molecules or vectors of the invention are provided.
  • Another aspect of the present invention thus relates to a non-human eukaryotic host strain expressing glycoproteins comprising modified N-glycans that resemble those made by human-cells. Performing the methods of the invention in species other than yeast and fungal cells is thus contemplated and encompassed by this invention. It is contemplated that a combinatorial nucleic acid library of the present invention may be used to select constructs that modify the glycosylation pathway in any eukaryotic host cell system. For example, the combinatorial libraries of the invention may also be used in plants, algae and insects, and in other eukaryotic host cells, including mammalian and human cells, to localize proteins, including glycosylation enzymes or catalytic domains thereof, in a desired location along a host cell secretory pathway. Preferably, glycosylation enzymes or catalytic domains and the like are targeted to a subcellular location along the host cell secretory pathway where they are capable of functioning, and preferably, where they are designed or selected to function most efficiently.
  • Plant and insect cells may also be engineered to alter the glycosylation of expressed proteins using the combinatorial library and methods of the invention. Furthermore, glycosylation in mammalian cells, including human cells, may also be modified using the combinatorial library and methods of the invention. It may be possible, for example, to optimize a particular enzymatic activity or to otherwise modify the relative proportions of various N-glycans made in a mammalian host cell using the combinatorial library and methods of the invention.
  • Examples of modifications to glycosylation which can be affected using a method according to this embodiment of the invention are: (1) engineering a eukaryotic host cell to trim mannose residues from Man8GlcNAc2 to yield a Man5GlcNAc2 N-glycan; (2) engineering eukaryotic host cell to add an N-acetylglucosamine (GlcNAc) residue to Man5GlcNAc2 by action of GlcNAc transferase I; (3) engineering.a eukaryotic host cell to functionally express an enzyme such as an N-acetylglucosaminyl Transferase (GnTI, GnTII, GnTIII, GnTIV, GnTV, GnTVI), mannosidase II, fucosyltransferase (FT), galactosyl tranferase (GalT) or a sialyltransferase (ST).
  • By repeating the method, increasingly complex glycosylation pathways can be engineered into a target host, such as a lower eukaryotic microorganism. In one preferred embodiment, the host organism is transformed two or more times with DNA libraries including sequences encoding glycosylation activities. Selection of desired phenotypes may be performed after each round of transformation or alternatively after several transformations have occurred. Complex glycosylation pathways can be rapidly engineered in this manner.
  • Sequential Glycosylation Reactions
  • In a preferred embodiment, such targeting peptide/catalytic domain libraries are designed to incorporate existing information on the sequential nature of glycosylation reactions in higher eukaryotes. Reactions known to occur early in the course of glycoprotein processing require the targeting of enzymes that catalyze such reactions to an early part of the Golgi or the ER. For example, the trimming of Man8GlcNAc2 to Man5GlcNAc2 by mannosidases is an early step in complex N-glycan formation (FIGS. 1B and 35A). Because protein processing is initiated in the ER and then proceeds through the early, medial and late Golgi, it is desirable to have this reaction occur in the ER or early Golgi. When designing a library for mannosidase I localization, for example, one thus attempts to match ER and early Golgi targeting signals with the catalytic domain of mannosidase I.
  • Generating Additional Sequence Diversity
  • The method of this embodiment is most effective when a nucleic acid, e.g., a DNA library transformed into the host contains a large diversity of sequences, thereby increasing the probability that at least one transformant will exhibit the desired phenotype. Single amino acid mutations, for example, may drastically alter the activity of glycoprotein processing enzymes (Romero et al. (2000) J. Biol. Chem. 275(15):11071-4). Accordingly, prior to transformation, a DNA library or a constituent sub-library may be subjected to one or more techniques to generate additional sequence diversity. For example, one or more rounds of gene shuffling, error prone PCR, in vitro mutagenesis or other methods for generating sequence diversity, may be performed to obtain a larger diversity of sequences within the pool of fusion constructs.
  • Expression Control Sequences
  • In addition to the open reading frame sequences described above, it is generally preferable to provide each library construct with expression control sequences, such as promoters, transcription terminators, enhancers, ribosome binding sites, and other functional sequences as may be necessary to ensure effective transcription and translation of the fusion proteins upon transformation of fusion constructs into the host organism.
  • Suitable vector components, e.g., selectable markers, expression control sequences (e.g., promoter, enhancers, terminators and the like) and, optionally, sequences required for autonomous replication in a host cell, are selected as a function of which particular host cell is chosen. Selection criteria for suitable vector components for use in a particular mammalian or a lower eukaryotic host cell are routine. Preferred lower eukaryotic host cells of the invention include Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium lucknowense, Fusarium sp. Fusarium gramineum, Fusarium venenatum and Neurospora crassa. Where the host is Pichia pastoris, suitable promoters include, for example, the AOX1, AOX2, GAPDH and P40 promoters.
  • Selectable Markers
  • It is also preferable to provide each construct with at least one selectable marker, such as a gene to impart drug resistance or to complement a host metabolic lesion. The presence of the marker is useful in the subsequent selection of transformants; for example, in yeast the URA3, HIS4, SUC2, G418, BLA, or SH BLE genes may be used. A multitude of selectable markers are known and available for use in yeast, fungi, plant, insect, mammalian and other eukaryotic host cells.
  • Transformation
  • The nucleic acid library is then transformed into the host organism. In yeast, any convenient method of DNA transfer may be used, such as electroporation, the lithium chloride method, or the spheroplast method. In filamentous fungi and plant cells, conventional methods include particle bombardment, electroporation and agrobacterium mediated transformation. To produce a stable strain suitable for high-density culture (e.g., fermentation in yeast), it is desirable to integrate the DNA library constructs into the host chromosome. In a preferred embodiment, integration occurs via homologous recombination, using techniques well-known in the art. For example, DNA library elements are provided with flanking sequences homologous to sequences of the host organism. In this manner, integration occurs at a defined site in the host genome, without disruption of desirable or essential genes.
  • In an especially preferred embodiment, library DNA is integrated into the site of an undesired gene in a host chromosome, effecting the disruption or deletion of the gene. For example, integration into the sites of the OCH1, MNN1, or MNN4 genes allows the expression of the desired library DNA while preventing the expression of enzymes involved in yeast hypermannosylation of glycoproteins. In other embodiments, library DNA may be introduced into the host via a nucleic acid molecule, plasmid, vector (e.g., viral or retroviral vector), chromosome, and may be introduced as an autonomous nucleic acid molecule or by homologous or random integration into the host genome. In any case, it is generally desirable to include with each library DNA construct at least one selectable marker gene to allow ready selection of host organisms that have been stably transformed. Recyclable marker genes such as ura3, which can be selected for or against, are especially suitable.
  • Screening and Selection Processes
  • After transformation of the host strain with the DNA library, transformants displaying a desired glycosylation phenotype are selected. Selection may be performed in a single step or by a series of phenotypic enrichment and/or depletion steps using any of a variety of assays or detection methods. Phenotypic characterization may be carried out manually or using automated high-throughput screening equipment. Commonly, a host microorganism displays protein N-glycans on the cell surface, where various glycoproteins are localized.
  • One may screen for those cells that have the highest concentration of terminal GlcNAc on the cell surface, for example, or for those cells which secrete the protein with the highest terminal GlcNAc content. Such a screen may be based on a visual method, like a staining procedure, the ability to bind specific terminal GlcNAc binding antibodies or lectins conjugated to a marker (such lectins are available from E.Y. Laboratories Inc., San Mateo, Calif.), the reduced ability of specific lectins to bind to terminal mannose residues, the ability to incorporate a radioactively labeled sugar in vitro, altered binding to dyes or charged surfaces, or may be accomplished by using a Fluorescence Assisted Cell Sorting (FACS) device in conjunction with a fluorophore labeled lectin or antibody (Guillen et al. (1998) Proc. Natl. Acad. Sci. USA 95(14):7888-7892).
  • Accordingly, intact cells may be screened for a desired glycosylation phenotype by exposing the cells to a lectin or antibody that binds specifically to the desired N-glycan. A wide variety of oligosaccharide-specific lectins are available commercially (e.g., from EY Laboratories, San Mateo, Calif.). Alternatively, antibodies to specific human or animal N-glycans are available commercially or may be produced using standard techniques. An appropriate lectin or antibody may be conjugated to a reporter molecule, such as a chromophore, fluorophore, radioisotope, or an enzyme having a chromogenic substrate (Guillen et al., 1998. Proc. Natl. Acad. Sci. USA 95(14): 7888-7892).
  • Screening may then be performed using analytical methods such as spectrophotometry, fluorimetry, fluorescence activated cell sorting, or scintillation counting. In other cases, it may be necessary to analyze isolated glycoproteins or N-glycans from transformed cells. Protein isolation may be carried out by techniques known in the art. In a preferred embodiment, a reporter protein is secreted into the medium and purified by affinity chromatography (e.g. Ni-affinity or glutathione-S-transferase affinity chromatography). In cases where an isolated N-glycan is preferred, an enzyme such as endo-β-N-acetylglucosaminidase (Genzyme Co., Boston, Mass.; New England Biolabs, Beverly, Mass.) may be used to cleave the N-glycans from glycoproteins. Isolated proteins or N-glycans may then be analyzed by liquid chromatography (e.g., HPLC), mass spectroscopy, or other suitable means. U.S. Pat. No. 5,595,900 teaches several methods by which cells with desired extracellular carbohydrate structures may be identified. In a preferred embodiment, MALDI-TOF mass spectrometry is used to analyze the cleaved N-glycans.
  • Prior to selection of a desired transformant, it may be desirable to deplete the transformed population of cells having undesired phenotypes. For example, when the method is used to engineer a functional mannosidase activity into cells, the desired transformants will have lower levels of mannose in cellular glycoprotein. Exposing the transformed population to a lethal radioisotope of mannose in the medium depletes the population of transformants having the undesired phenotype, i.e., high levels of incorporated mannose (Huffaker and Robbins (1983) Proc Natl Acad Sci USA. 80(24):7466-70). Alternatively, a cytotoxic lectin or antibody, directed against an undesirable N-glycan, may be used to deplete a transformed population of undesired phenotypes (e.g., Stanley and Siminovitch (1977) Somatic Cell Genet 3(4):391-405). U.S. Pat. No. 5,595,900 teaches several methods by which cells with a desired extracellular carbohydrate structures may be identified. Repeatedly carrying out this strategy allows for the sequential engineering of more and more complex glycans in lower eukaryotes.
  • To detect host cells having on their surface a high degree of the human-like N-glycan intermediate GlcNAcMan3GlcNAc2, for example, one may select for transformants that allow for the most efficient transfer of GlcNAc by GlcNAc Transferase from UDP-GlcNAc in an in vitro cell assay. This screen may be carried out by growing cells harboring the transformed library under selective pressure on an agar plate and transferring individual colonies into a 96-well microtiter plate. After growing the cells, the cells are centrifuged, the cells resuspended in buffer, and after addition of UDP-GlcNAc and GnTII, the release of UDP is determined either by HPLC or an enzyme linked assay for UDP. Alternatively, one may use radioactively labeled UDP-GlcNAc and GnTII, wash the cells and then look for the release of radioactive GlcNAc by N-actylglucosaminidase. All this may be carried manually or automated through the use of high throughput screening equipment. Transformants that release more UDP, in the first assay, or more radioactively labeled GlcNAc in the second assay, are expected to have a higher degree of GlcNAcMan3GlcNAc2 on their surface and thus constitute the desired phenotype. Similar assays may be adapted to look at the N-glycans on secreted proteins as well.
  • Alternatively, one may use any other suitable screen such as a lectin binding assay that is able to reveal altered glycosylation patterns on the surface of transformed cells. In this case the reduced binding of lectins specific to terminal mannoses may be a suitable selection tool. Galantus nivalis lectin binds specifically to terminal α-1,3 mannose, which is expected to be reduced if sufficient mannosidase II activity is present in the Golgi. One may also enrich for desired transformants by carrying out a chromatographic separation step that allows for the removal of cells containing a high terminal mannose content. This separation step would be carried out with a lectin column that specifically binds cells with a high terminal mannose content (e.g., Galantus nivalis lectin bound to agarose, Sigma, St.Louis, Mo.) over those that have a low terminal mannose content.
  • In addition, one may directly create such fusion protein constructs, as additional information on the localization of active carbohydrate modifying enzymes in different lower eukaryotic hosts becomes available in the scientific literature. For example, it is known that human β1,4-GalTr can be fused to the membrane domain of MNT, a mannosyltransferase from S. cerevisiae, and localized to the Golgi apparatus while retaining its catalytic activity (Schwientek et al. (1995) J. Biol. Chem. 270(10):5483-9). If S. cerevisiae or a related organism is the host to be engineered one may directly incorporate such findings into the overall strategy to obtain complex N-glycans from such a host. Several such gene fragments in P. pastoris have been identified that are related to glycosyltransferases in S. cerevisiae and thus could be used for that purpose.
  • Integration Sites
  • As one ultimate goal of this genetic engineering effort is a robust protein production strain that is able to perform well in an industrial fermentation process, the integration of multiple genes into the host (e.g., fungal) chromosome preferably involves careful planning. The engineered strain may likely have to be transformed with a range of different genes, and these genes will have to be transformed in a stable fashion to ensure that the desired activity is maintained throughout the fermentation process. As described herein, any combination of various desired enzyme activities may be engineered into the fungal protein expression host, e.g., sialyltransferases, mannosidases, fucosyltransferases, galactosyltransferases, glucosyltransferases, GlcNAc transferases, ER and Golgi specific transporters (e.g. syn and antiport transporters for UDP-galactose and other precursors), other enzymes involved in the processing of oligosaccharides, and enzymes involved in the synthesis of activated oligosaccharide precursors such as UDP-galactose, CMP-N-acetylneuraminic acid. Examples of preferred methods for modifying glycosylation in a lower eukaryotic host cell, such as Pichia pastoris, are shown in Table 6.
    TABLE 6
    Some preferred embodiments for modifying glycosylation in a lower eukaroytic microorganism
    Suitable
    Suitable Suitable Sources of Suitable Transporters
    Desired Catalytic Localization Gene and/or
    Structure Activities Sequences Deletions Phosphatases
    Man5GlcNAc2 α-1,2- Mns1 (N-terminus, OCH1 none
    mannosidase S. cerevisiae) MNN4
    (murine, Och1 (N-terminus, MNN6
    human, S. cerevisiae, P. pastoris)
    Bacillus sp., Ktr1
    A. nidulans) Mnn9
    Mnt1 (S. cerevisiae)
    KDEL, HDEL
    (C-terminus)
    GlcNAcMan5GlcNAc2 GlcNAc Och1 (N-terminus, OCH1 UDP-GlcNAc
    Transferase I, S. cerevisiae, P. pastoris) MNN4 transporter
    (human, KTR1 (N-terminus) MNN6 (human, murine,
    murine, rat Mnn1 (N-terminus, K. lactis)
    etc.) S. cerevisiae) UDPase
    Mnt1 (N-terminus, (human)
    S. cerevisiae)
    GDPase
    (N-terminus, S. cerevisiae)
    GlcNAcMan3GlcNAc2 mannosidase Ktr1 OCH1 UDP-GlcNAc
    II Mnn1 (N-terminus, MNN4 transporter
    S. cerevisiae) MNN6 (human, murine,
    Mnt1(N-terminus, S. cerevisiae) K. lactis)
    Kre2/Mnt1 (S. cerevisiae) UDPase
    Kre2 (P. pastoris) (human)
    Ktr1 (S. cerevisiae)
    Ktr1 (P. pastoris)
    Mnn1 (S. cerevisiae)
    GlcNAc(2-4)- GlcNAc Mnn1 (N-terminus, OCH1 UDP-GlcNAc
    Man3GlcNAc2 Transferase S. cerevisiae) MNN4 transporter
    II, III, IV, V Mnt1 (N-terminus, MNN6 (human, murine,
    (human, S. cerevisiae) K. lactis)
    murine) Kre2/Mnt1 (S. cerevisiae) UDPase
    Kre2 (P. pastoris) (human)
    Ktr1 (S. cerevisiae)
    Ktr1 (P. pastoris)
    Mnn1 (S. cerevisiae)
    Gal(1-4)GlcNAc(2-4)- β-1,4- Mnn1 (N-terminus, OCH1 UDP-Galactose
    Man3GlcNAc2 Galactosyl S. cerevisiae) MNN4 transporter
    transferase Mnt1(N-terminus, S. cerevisiae) MNN6 (human,
    (human) Kre2/Mnt1 (S. cerevisiae) S. pombe)
    Kre2 (P. pastoris)
    Ktr1 (S. cerevisiae)
    Ktr1 (P. pastoris)
    Mnn1 (S. cerevisiae)
    NANA(1-4)- α-2,6- KTR1 OCH1 CMP-Sialic acid
    Gal(1-4)GlcNAc(2-4)- Sialyltransferase MNN1 (N-terminus, MNN4 transporter
    Man3GlcNAc2 (human) S. cerevisiae) MNN6 (human)
    α-2,3- MNT1 (N-terminus,
    Sialyltransferase S. cerevisiae)
    Kre2/Mnt1 (S. cerevisiae)
    Kre2 (P. pastoris)
    Ktr1 (S. cerevisiae)
    Ktr1 (P. pastoris)
    MNN1 (S. cerevisiae)
  • As any strategy to engineer the formation of complex N-glycans into a host cell such as a lower eukaryote involves both the elimination as well as the addition of particular glycosyltransferase activities, a comprehensive scheme will attempt to coordinate both requirements. Genes that encode enzymes that are undesirable serve as potential integration sites for genes that are desirable. For example, 1,6 mannosyltransferase activity is a hallmark of glycosylation in many known lower eukaryotes. The gene encoding alpha-1,6 mannosyltransferase (OCH1) has been cloned from S. cerevisiae and mutations in the gene give rise to a viable phenotype with reduced mannosylation. The gene locus encoding alpha-1,6 mannosyltransferase activity therefore is a prime target for the integration of genes encoding glycosyltransferase activity. In a similar manner, one can choose a range of other chromosomal integration sites that, based on a gene disruption event in that locus, are expected to: (1) improve the cells ability to glycosylate in a more human-like fashion, (2) improve the cells ability to secrete proteins, (3) reduce proteolysis of foreign proteins and (4) improve other characteristics of the process that facilitate purification or the fermentation process itself.
  • Target Glycoproteins
  • The methods described herein are useful for producing glycoproteins, especially glycoproteins used therapeutically in humans. Glycoproteins having specific glycoforms may be especially useful, for example, in the targeting of therapeutic proteins. For example, mannose-6-phosphate has been shown to direct proteins to the lysosome, which may be essential for the proper function of several enzymes related to lysosomal storage disorders such as Gaucher's, Hunter's, Hurler's, Scheie's, Fabry's and Tay-Sachs disease, to mention just a few. Likewise, the addition of one or more sialic acid residues to a glycan side chain may increase the lifetime of a therapeutic glycoprotein in vivo after administration. Accordingly, host cells (e.g., lower eukaryotic or mammalian) may be genetically engineered to increase the extent of terminal sialic acid in glycoproteins expressed in the cells. Alternatively, sialic acid may be conjugated to the protein of interest in vitro prior to administration using a sialic acid transferase and an appropriate substrate. Changes in growth medium composition may be employed in addition to the expression of enzyme activities involved in human-like glycosylation to produce glycoproteins more closely resembling human forms (Weikert et al. (1999) Nature Biotechnology 17, 1116-1121; Werner et al. (1998) Arzneimittelforschung 48(8):870-880; Andersen and Goochee (1994) Cur. Opin. Biotechnol. 5:546-549; Yang and Butler (2000) Biotechnol.Bioengin. 68(4):370-380). Specific glycan modifications to monoclonal antibodies (e.g. the addition of a bisecting GlcNAc) have been shown to improve antibody dependent cell cytotoxicity (Umana et al. (1999) Nat. Biotechnol. 17(2):176-80), which may be desirable for the production of antibodies or other therapeutic proteins.
  • Therapeutic proteins are typically administered by injection, orally, pulmonary, or other means. Examples of suitable target glycoproteins which may be produced according to the invention include, without limitation: erythropoietin, cytokines such as interferon-α, interferon-β, interferon-γ, interferon-ω, and granulocyte-CSF, coagulation factors such as factor VIII, factor IX, and human protein C, soluble IgE receptor α-chain, IgG, IgG fragments, IgM, interleukins, urokinase, chymase, urea trypsin inhibitor, IGF-binding protein, epidermal growth factor, growth hormone-releasing factor, annexin V fusion protein, angiostatin, vascular endothelial growth factor-2, myeloid progenitor inhibitory factor-1, osteoprotegerin, α-1-antitrypsin, Dnase II, and α-feto proteins.
  • Expression Of GnT-III To Boost Antibody Functionality
  • The addition of N-acetylglucosamine residues to the GlcNAcMan3GlcNAc2 structure by N-acetylglucosaminyltransferases II and III yields a so-called bisected N-glycan GlcNAc3Man3GlcNAc2 (FIG. 15). This structure has been implicated in greater antibody-dependent cellular cytotoxicity (ADCC) (Umana et al. (1999) Nat. Biotechnol. 17(2):176-80). Re-engineering glycoforms of immunoglobulins expressed by mammalian cells is a tedious and cumbersome task. Especially in the case of GnTIII, where over-expression of this enzyme has been implicated in growth inhibition, methods involving regulated (inducible) gene expression had to be employed to produce immunoglobulins with bisected N-glycans (Umana et al. (1999) Biotechnol Bioeng. 65(5):542-9; Umana et al. (1999) Nat. Biotechnol. 17(2):176-80); Umana et al. WO 03/011878; U.S. Pat. No. 6,602,684.
  • Accordingly, in another embodiment, the invention provides systems and methods for producing human-like N-glycans having bisecting N-acetylglucosamine (GlcNAc) on a trimannose or pentamannose core structure. In a preferred embodiment, the invention provides a system and method for producing immunoglobulins having bisected N-glycans. The systems and methods described herein will not suffer from previous problems, e.g., cytotoxicity associated with overexpression of GnTIII or ADCC, as the host cells of the invention are engineered and selected to be viable and preferably robust cells which produce N-glycans having substantially modified human-type glycoforms such as GlcNAc2Man3GlcNAc2. Thus, addition of a bisecting N-acetylglucosamine in a host cell of the invention will have a negligible effect on the growth-phenotype or viability of those host cells.
  • In addition, work by others has shown that there is no linear correlation between GnTIII expression levels and the degree of ADCC. Umana et al. (1999) Nature Biotechnol. 17:176-80. Thus, finding the optimal expression level in mammalian cells and maintaining it throughout an FDA approved fermentation process seems to be a challenge. However, in cells of the invention, such as fungal cells, finding a promoter of appropriate strength to establish a robust, reliable and optimal GnTIII expression level is a comparatively easy task for one of skill in the art.
  • A host cell such as a yeast strain capable of producing glycoproteins with bisecting N-glycans is engineered according to the invention, by introducing into the host cell a GnTIII activity (Example 12). Preferably, the host cell is transformed with a nucleic acid that encodes GnTIII (see, e.g., FIG. 24) or a domain thereof having enzymatic activity, optionally fused to a heterologous cell signal targeting peptide (e.g., using the libraries and associated methods of the invention.) Host cells engineered to express GnTIII will produce higher antibody titers than mammalian cells are capable of. They will also produce antibodies with higher potency with respect to ADCC.
  • Antibodies produced by mammalian cell lines transfected with GnTIII have been shown to be as effective as antibodies produced by non-transfected cell-lines, but at a 10-20 fold lower concentration (Davies et al. (2001) Biotechnol. Bioeng. 74(4):288-94). An increase of productivity of the production vehicle of the invention over mammalian systems by a factor of twenty, and a ten-fold increase of potency will result in a net-productivity improvement of two hundred. The invention thus provides a system and method for producing high titers of an antibody having high potency (e.g., up to several orders of magnitude more potent than what can currently be produced). The system and method is safe and provides high potency antibodies at low cost in short periods of time. Host cells engineered to express GnTIII according to the invention produce immunoglobulins having bisected N-glycans at rates of at least 50 mg/liter/day to at least 500 mg/liter/day. In addition, each immunoglobulin (Ig) molecule (comprising bisecting GlcNAcs) is more potent than the same Ig molecule produced without bisecting GlcNAcs.
  • The following are examples which illustrate various aspects of the invention. These examples should not be construed as limiting: the examples are included for the purposes of illustration only.
  • EXAMPLE 1 Cloning and Disruption of the OCH1 gene in P. pastoris
  • Generation of an OCH1 mutant of P. pastoris:
  • A 1215 bp ORF of the P. pastoris OCH1 gene encoding a putative α-1,6 mannosyltransferase was amplified from P. pastoris genomic DNA (strain X-33, Invitrogen, Carlsbad, Calif.) using the oligonucleotides 5′-ATGGCGAAGGCAGATGGCAGT-3′ (SEQ ID NO:3) and 5′-TTAGTCCTTCCAACTTCCTTC-3′ (SEQ ID NO:4) which were designed based on the P. pastoris OCH1 sequence (Japanese Patent Application Publication No. 8-336387). Subsequently, 2685 bp upstream and 1175 bp downstream ofthe ORF of the OCH1 gene were amplified from a P. pastoris genomic DNA library (Boehm, T. et al. (1999) Yeast 15(7):563-72) using the internal oligonucleotides 5′-ACTGCCATCTGCCTTCGCCAT-3′ (SEQ ID NO:47) in the OCH1 gene, and 5′-GTAATACGACTCACTATAGGGC-3′ T7 (SEQ ID NO:48) and 5′-AATTAACCCTCACTAAAGGG-3′ T3 (SEQ ID NO:49) oligonucleotides in the backbone of the library bearing plasmid lambda ZAP II (Stratagene, La Jolla, Calif.). The resulting 5075 bp fragment was cloned into the pCR2.1-TOPO vector (Invitrogen, Carlsbad, Calfi.) and designated pBK9.
  • After assembling a gene knockout construct that substituted the OCH1 reading frame with a HIS4 resistance gene, P. pastoris was transformed and colonies were screened for temperature sensitivity at 37° C. OCH1 mutants of S. cerevisiae are temperature sensitive and are slow growers at elevated temperatures. One can thus identify functional homologs of OCH1 in P. pastoris by complementing an OCH1 mutant of S. cerevisiae with a P. pastoris DNA or cDNA library. About 20 temperature sensitive strains were further subjected to a colony PCR screen to identify colonies with a deleted och1 gene. Several och1 deletions were obtained.
  • The linearized pBK9.1, which has 2.1 kb upstream sequence and 1.5 kb downstream sequence of OCH1 gene cassette carrying Pichia HIS4 gene, was transformed into P. pastoris BK1 [GS1 15 (his4 Invitrogen Corp., San Diego, Calif.) carrying the human IFN-β gene in the AOX1 locus] to knock out the wild-type OCH1 gene. The initial screening of transformants was performed using histidine drop-out medium followed by replica plating to select the temperature sensitive colonies. Twenty out of two hundred histidine-positive colonies showed a temperature sensitive phenotype at 37° C. To exclude random integration of pBK9.1 into the Pichia genome, the 20 temperature-sensitive isolates were subjected to colony PCR using primers specific to the upstream sequence of the integration site and to HIS4 ORF. Two out of twenty colonies were ochl defective and further analyzed using a Southern blot and a Western blot indicating the functional och1 disruption by the och1 knock-out construct. Genomic DNA were digested using two separate restriction enzymes BglII and ClaI to confirm the och1 knock-out and to confirm integration at the open reading frame. The Western Blot showed och1 mutants lacking a discrete band produced in the GS 115 wild type at 46.2 kDa.
  • EXAMPLE 2 Engineering of P. pastoris with α-1,2-Mannosidase to Produce Man5GlcNAc2-Containing IFN-β Precursors
  • An α-1,2-mannosidase is required for the trimming of Man8GlcNAc2 to yield Man5GlcNAc2, an essential intermediate for complex N-glycan formation. While the production of a Man5GlcNAc2 precursor is essential, it is not necessarily sufficient for the production of hybrid and complex glycans because the specific isomer of Man5GlcNAc2 may or may not be a substrate for GnTI. An och1 mutant of P. pastoris is engineered to express secreted human interferon-β under the control of an aox promoter. A DNA library is constructed by the in-frame ligation of the catalytic domain of human mannosidase IB (an α-1,2-mannosidase) with a sub-library including sequences encoding early Golgi and ER localization peptides. The DNA library is then transformed into the host organism, resulting in a genetically mixed population wherein individual transformants each express interferon-β as well as a synthetic mannosidase gene from the library. Individual transformant colonies are cultured and the production of interferon is induced by addition of methanol. Under these conditions, over 90% of the secreted protein is glycosylated interferon-β.
  • Supernatants are purified to remove salts and low-molecular weight contaminants by C18 silica reversed-phase chromatography. Desired transformants expressing appropriately targeted, active α-1,2-mannosidase produce interferon-β including N-glycans of the structure Man5GlcNAc2, which has a reduced molecular mass compared to the interferon-β of the parent strain. The purified interferon-β is analyzed by MALDI-TOF mass spectroscopy and colonies expressing the desired form of interferon-β are identified.
  • EXAMPLE 3 Generation of an och1 Mutant Strain Expressing an α-1,2-Mannosidase, GnTI and GnTII for Production of a Human-Like Glycoprotein
  • The 1215 bp open reading frame of the P. pastoris OCH1 gene as well as 2685 bp upstream and 1175 bp downstream was amplified by PCR (see also WO 02/00879), cloned into the pCR2.1-TOPO vector (Invitrogen) and designated pBK9. To create an och1 knockout strain containing miultiple auxotrophic markers, 100 μg of pJN329, a plasmid containing an och1:: URA3 mutant allele flanked with SfiI restriction sites was digested with SfiI and used to transform P. pastoris strain JC308 (Cereghino et al. (2001) Gene 263:159-169) by electroporation. Following incubation on defined medium lacking uracil for 10 days at room temperature, 1000 colonies were picked and re-streaked. URA+ clones that were unable to grow at 37° C., but grew at room temperature, were subjected to colony PCR to test for the correct integration of the och1:: URA3 mutant allele. One clone that exhibited the expected PCR pattern was designated YJN153. The Kringle 3 domain of human plasminogen (K3) was used as a model protein. A NeoR marked plasmid containing the K3 gene was transformed into strain YJN153 and a resulting strain, expressing K3, was named BK64-1.
  • Plasmid pPB103, containing the Kluyveromyces lactis MNN2-2 gene which encodes a Golgi UDP-N-acetylglucosamine transporter was constructed by cloning a blunt BglII-HindIII fragment from vector pDL02 (Abeijon et al. (1996) Proc. Natl. Acad. Sci. U.S.A. 93:5963-5968) into BgllI and BamHI digested and blunt ended pBLADE-SX containing the P. pastoris ADE1 gene (Cereghino et al. (2001) Gene 263:159-169). This plasmid was linearized with EcoNI and transformed into strain BK64-1 by electroporation and one strain confirmed to contain the MNN2-2 by PCR analysis was named PBP1.
  • A library of mannosidase constructs was generated, comprising in-frame fusions of the leader domains of several type I or type II membrane proteins from S. cerevisiae and P. pastoris fused with the catalytic domains of several α-1,2-mannosidase genes from human, mouse, fly, worm and yeast sources (see, e.g., WO02/00879, incorporated herein by reference). This library was created in a P. pastoris HIS4 integration vector and screened by linearizing with SalI, transforming by electroporation into strain PBP1, and analyzing the glycans released from the K3 reporter protein. One active construct chosen was a chimera of the 988-1296 nucleotides (C-terminus) of the yeast SEC12 gene fused with a N-terminal deletion of the mouse α-1,2-mannosidase IA gene (FIG. 3), which was missing the 187 nucleotides. A P. pastoris strain expressing this construct was named PBP2.
  • A library of GnTI constructs was generated, comprising in-frame fusions of the same leader library with the catalytic domains of GnTI genes from human, worm, frog and fly sources (WO 02/00879). This library was created in a P. pastoris ARG4 integration vector and screened by linearizing with AatII, transforming by electroporation into strain PBP2, and analyzing the glycans released from K3. One active construct chosen was a chimera of the first 120 bp of the S. cerevisiae MNN9 gene fused to a deletion of the human GnTI gene, which was missing the first 154 bp. A P. pastoris strain expressing this construct was named PBP-3. (See also FIG. 36.)
  • A library of GnTII constructs was generated, which comprised in-frame fusions of the leader library with the catalytic domains of GnTII genes from human and rat sources (WO 02/00879). This library was created in a P. pastoris integration vector containing the NSTR gene conferring resistance to the drug nourseothricin. The library plasmids were linearized with EcoRI, transformed into strain RDP27 by electroporation, and the resulting strains were screened by analysis of the released glycans from purified K3.
  • Materials for the Following Reactions
  • MOPS, sodium cacodylate, manganese chloride, UDP-galactose and CMP-N-acetylneuraminic acid were from Sigma. Trifluoroacetic acid (TFA) was from Sigma/Aldrich, Saint Louis, Mo. Recombinant rat α2,6-sialyltransferase from Spodoptera frugiperda and β1,4-galactosyltransferase from bovine milk were from Calbiochem (San Diego, Calif.). Protein N-glycosidase F, mannosidases, and oligosaccharides were from Glyko (San Rafael, Calif.). DEAE ToyoPearl resin was from TosoHaas. Metal chelating “HisBind” resin was from Novagen (Madison, Wis.). 96-well lysate-clearing plates were from Promega (Madison, Wis.). Protein-binding 96-well plates were from Millipore (Bedford, Mass.). Salts and buffering agents were from Sigma (St. Louis, Mo.). MALDI matrices were from Aldrich (Milwaukee, Wis.).
  • Protein Purification
  • Kringle 3 was purified using a 96-well format on a Beckman BioMek 2000 sample-handling robot (Beckman/Coulter Ranch Cucamonga, Calif.). Kringle 3 was purified from expression media using a C-terminal hexa-histidine tag. The robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin. Briefly, a 150 uL (μL) settled volume of resin is poured into the wells of a 96-well lysate-binding plate, washed with 3 volumes of water and charged with 5 volumes of 50 mM NiSO4 and washed with 3 volumes of binding buffer (5 mM imidazole, 0.5 M NaCl, 20 mM Tris-HCL pH7.9). The protein expression media is diluted 3:2, media/PBS (60 mM PO4, 16 mM KCl, 822 mM NaCl pH7.4) and loaded onto the columns. After draining, the columns are washed with 10 volumes of binding buffer and 6 volumes of wash buffer (30 mM imidazole, 0.5 M NaCl, 20 mM Tris-HCl pH7.9) and the protein is eluted with 6 volumes of elution buffer (1 M imidazole, 0.5 M NaCl, 20 mM Tris-HCl pH7.9). The eluted glycoproteins are evaporated to dryness by lyophilyzation.
  • Release of N-linked Glycans
  • The glycans are released and separated from the glycoproteins by a modification of a previously reported method (Papac, et al. A. J. S. (1998) Glycobiology 8, 445-454). The wells of a 96-well MultiScreen IP (Immobilon-P membrane) plate (Millipore) are wetted with 100 uL of methanol, washed with 3×150 uL of water and 50 uL of RCM buffer (8M urea, 360 mM Tris, 3.2 mM EDTA pH8.6), draining with gentle vacuum after each addition. The dried protein samples are dissolved in 30 uL of RCM buffer and transferred to the wells containing 10 uL of RCM buffer. The wells are drained and washed twice with RCM buffer. The proteins are reduced by addition of 60 uL of 0.1 M DTT in RCM buffer for 1 hr at 37° C. The wells are washed three times with 300 uL of water and carboxymethylated by addition of 60 uL of 0.1 M iodoacetic acid for 30min in the dark at room temperature. The wells are again washed three times with water and the membranes blocked by the addition of 100 uL of 1% PVP 360 in water for 1 hr at room temperature. The wells are drained and washed three times with 300 uL of water and deglycosylated by the addition of 30 uL of 10 mM NH4HCO3 pH 8.3 containing one milliunit of N-glycanase (Glyko). After 16 hours at 37° C., the solution containing the glycans was removed by centrifugation and evaporated to dryness.
  • Matrix Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry
  • Molecular weights of the glycans were determined using a Voyager DE PRO linear MALDI-TOF (Applied Biosciences) mass spectrometer using delayed extraction. The dried glycans from each well were dissolved in 15 uL of water and 0.5 uL spotted on stainless steel sample plates and mixed with 0.5 uL of S-DHB matrix (9 mg/mL of dihydroxybenzoic acid, 1 mg/mL of 5-methoxysalicilic acid in 1:1 water/acetonitrile 0.1% TFA) and allowed to dry.
  • Ions were generated by irradiation with a pulsed nitrogen laser (337 nm) with a 4 ns pulse time. The instrument was operated in the delayed extraction mode with a 125 ns delay and an accelerating voltage of 20 kV. The grid voltage was 93.00%, guide wire voltage was 0.10%, the internal pressure was less than 5×10-7 torr, and the low mass gate was 875 Da. Spectra were generated from the sum of 100-200 laser pulses and acquired with a 2 GHz digitizer. Man5GlcNAc2 oligosaccharide was used as an external molecular weight standard. All spectra were generated with the instrument in the positive ion mode. The estimated mass accuracy of the spectra was 0.5%.
  • EXAMPLE 4 Engineering of P. pastoris to Produce Man5GlcNAc2 as the Predominant N-Glycan Structure Using a Combinatorial DNA Library
  • An och1 mutant of P. pastoris (see Examples 1 and 3) was engineered to express and secrete proteins such as the kringle 3 domain of human plasminogen (K3) under the control of the inducible AOXI promoter. The Kringle 3 domain of human plasminogen (K3) was used as a model protein. A DNA fragment encoding the K3 was amplified using Pfu turbo polymerase (Strategene, La Jolla, Calif.) and cloned into EcoRI and XbaI sites of pPICZaA (Invitrogen, Carlsbad, Calif.), resulting in a C-terminal 6- His tag. In order to improve the N-linked glycosylation efficiency of K3 (Hayes et al. 1975 J Arch. Biochem. Biophys. 171, 651-655), Pro46 was replaced with Ser46 using site-directed mutagenesis. The resulting plasmid was designated pBK64. The correct sequence of the PCR construct was confirmed by DNA sequencing.
  • A combinatorial DNA library was constructed by the in-frame ligation of murine α-1,2-mannosidase IB (Genbank AN 6678787) and IA (Genbank AN 6754619) catalytic domains with a sub-library including sequences encoding Cop II vesicle, ER, and early Golgi localization peptides according to Table 6. The combined DNA library was used to generate individual fusion constructs, which were then transformed into the K3 expressing host organism, resulting in a genetically mixed population wherein individual transformants each express K3 as well as a localization signal/mannosidase fusion gene from the library. Individual transform ants were cultured and the production of K3 was induced by transfer to a methanol containing medium. Under these conditions, after 24 hours of induction, over 90% of the protein in the medium was K3. The K3 reporter protein was purified from the supernatant to remove salts and low-molecular weight contaminants by Ni-affinity chromatography. Following affinity purification, the protein was desalted by size exclusion chromatography on a Sephadex G10 resin (Sigma, St. Louis, Mo.) and either directly subjected to MALDI-TOF analysis described below or the N-glycans were removed by PNGase digestion as described below (Release of N-glycans) and subjected to MALDI-TOF analysis Miele et al. (1997) Biotechnol. Appl. Biochem. 25:151-157.
  • Following this approach, a diverse set of transformants were obtained; some showed no modification of the N-glycans compared to the och1 knockout strain; and others showed a high degree of mannose trimming (FIGS. 5D and 5E). Desired transformants expressing appropriately targeted, active α-1,2-mannosidase produced K3 with N-glycans of the structure Man5GlcNAc2. This confers a reduced molecular mass to the glycoprotein compared to the K3 of the parent och1 deletion strain, a difference which was readily detected by MALDI-TOF mass spectrometry (FIG. 5). Table 7 indicates the relative Man5GlcNAc2 production levels.
    TABLE 7
    A representative combinatorial DNA library of localization
    sequences/catalytic domains exhibiting relative levels of Man5GlcNAc2
    production.
    Targeting peptide sequences
    MNS1 (s) MNS1 (m) MNS1 (I) SEC12 (s) SEC12 (m)
    Catalytic Domians Mouse mannosidase FB4 FB5 FB6 FB7 FB8
    1A Δ187 ++ + ++ ++++
    Mouse mannosidase GB4 GB5 GB6 GB7 GB8
    1B Δ58 ++ + + ++ +
    Mouse mannosidase GC4 GC5 GC6 GC7 GC8
    1B Δ99 +++ + + +
    Mouse mannosidase GD4 GD5 GD6 GD7 GD8
    1B Δ170 + +
  • TABLE 8
    Another combinatorial DNA library of localization
    sequences/catalytic domains exhibiting relative levels of Man5GlcNAc2
    production.
    Targeting peptide sequences
    VAN1 (s) VAN1 (m) VANi (I) MNN10 (s) MNN10 (m) MNN10 (I)
    Catalytic Domians C. elegans BC18-5 BC19 BC20 BC27 BC28 BC29
    mannosidase +++++ ++++ +++ +++++ +++++ +++
    1B Δ80
    C. elegans BB18 BB19 BB20 BB18 BB19 BB20
    mannosidase +++++ +++++ ++++ +++++ +++++ ++++
    1B Δ31
  • Targeting peptides were selected from MNS I (SwissProt P32906) in S. cerevisiae (long, medium and short) (see supra Nucleic Acid Libraries; Combinatorial DNA Library of Fusion Constructs) and SEC12 (SwissProt P11655) in S. cerevisiae (988-1140 nucleotides: short) and (988-1296: medium). Although majority of the targeting peptide sequences were N-terminal deletions, some targeting peptide sequences, such as SEC12 were C-terminal deletions. Catalytic domains used in this experiment were selected from mouse mannosidase 1A with a 187 amino acid N-terminal deletion; and mouse mannosidase 1B with a 58, 99 and 170 amino acid deletion. The number of (+)s, as used herein, indicates the relative levels of Man5GlcNAc2 production. The notation (−) indicates no apparent production of Man5GlcNAc2. The notation (+) indicates less than 10% production of Man5GlcNAc2 The notation (++) indicates about 10-20% production of Man5GlcNAc2. The notation with (+++) indicates about 20-40% production of Man5GlcNAc2. The notation with (++++) indicates about 50% production of Man5GlcNAc2. The notation with (+++++) indicates greater than 50% production of Man5GlcNAC2.
  • Table 9 shows relative amount of Man5GlcNAc2 on secreted K3. Six hundred and eight (608) different strains of P. pastoris, Δoch1 were generated by transforming them with a single construct of a combinatorial genetic library that was generated by fusing nineteen (19) α-1,2 mannosidase catalytic domains to thirty-two (32) fungal ER, and cis-Golgi leaders.
    TABLE 9
    Amount of Man5GlcNAc2 on Number of
    secreted K3 (% of total glycans) constructs (%)
    N.D.*  19 (3.1)
     0-10% 341 (56.1)
    10-20%  50 (8.2)
    20-40&  75 (12.3)
    40-60%  72 (11.8)
    More than 60%  51 (8.4)
    Total 608 (100)

    *Several fusion constructs were not tested because the corresponding plasmids could not be propagated in E. coli prior to transformation into P. pastoris.

    Clones with the highest degree of Man5GlcNAc2 trimming (30/51) were further analyzed for mannosidase activity in the supernatant of the medium. The majority (28/30) displayed detectable mannosidase activity in the supernatant (e.g. FIG. 4B). Only two constructs displayed high Man5GlcNAc2 levels, while lacking mannosidase activity in the medium (e.g. FIG. 4C).
  • Table 7 shows two constructs pFB8 and pGC5, among others, displaying Man5GlcNAc2. Table 8 shows a more preferred construct, pBC18-5, a S. cerevisiae VAN1 (S) targeting peptide sequence (from SwissProt 23642) ligated in-frame to a C. elegans mannosidase IB (Genbank AN CAA98114) 80 amino acid N-terminal deletion (Saccharomyces Van1 (s)/C. elegans mannosidase IB Δ80). This fusion construct also produces a predominant Man5GlcNAc2 structure, as shown in FIG. 5E. This construct was shown to produce greater than 50% Man5GlcNAc2 (+++++).
  • Generation of a Combinatorial Localization/Mannosidase Library:
  • Generating a combinatorial DNA library of α-1,2-mannosidase catalytic domains fused to targeting peptides required the amplification of mannosidase domains with varying lengths of N-terminal deletions from a number of organisms. To approach this goal, the full length open reading frames (ORFs) of α-1,2-mannosidases were PCR amplified from either cDNA or genomic DNA obtained from the following sources: Homo sapiens, Mus musculus, Drosophila melanogaster, Caenorhabditis elegans, Aspergillus nidulans and Penicillium citrinum. In each case, DNA was incubated in the presence of oligonucleotide primers specific for the desired mannosidase sequence in addition to reagents required to perform the PCR reaction. For example, to amplify the ORF of the M. musculus α-1,2-mannosidase IA, the 5′-primer ATGCCCGTGGGGGGCCTGTTGCCGCTCTTCAGTAGC (SEQ ID NO:52) and the 3′-primer TCATTTCTCTTTGCCATCAATTTCCTTCTTCTGTTCACGG (SEQ ID NO:53) were incubated in the presence of Pfu DNA polymerase (Stratagene, La Jolla, Calif.) and amplified under the conditions recommended by Stratagene using the cycling parameters: 94° C. for 1 min (1 cycle); 94° C. for 30 sec, 68° C. for 30 sec, 72° C. for 3 min (30 cycles). Following amplification the DNA sequence encoding the ORF was incubated at 72° C. for 5 min with 1 U Taq DNA polymerase (Promega, Madison, Wis.) prior to ligation into pCR2.1 -TOPO (Invitrogen, Carlsbad, Calif.) and transformed into TOP 10 chemically competent E. coli, as recommended by Invitrogen. The cloned PCR product was confirmed by ABI sequencing using primers specific for the mannosidase ORF.
  • To generate the desired N-terminal truncations of each mannosidase, the complete ORF of each mannosidase was used as the template in a subsequent round of PCR reactions wherein the annealing position of the 5′-primer was specific to the 5′-terminus of the desired truncation and the 3′-primer remained specific for the original 3′-terminus of the ORF. To facilitate subdloning of the truncated mannosidase fragment into the yeast expression vector, pJN347 (FIG. 2C) AscI and Pacl restriction sites were engineered onto each truncation product, at the 5′- and 3′-termini respectively. The number and position of the N-terminal truncations generated for each mannosidase ORF depended on the position of the transmembrane (TM) region in relation to the catalytic domain (CD). For instance, if the stem region located between the TM and CD was less than 150 bp, then only one truncation for that protein was generated. If, however, the stem region was longer than 150 bp then either one or two more truncations were generated depending on the length of the stem region.
  • An example of how truncations for the M musculus mannosidase IA (Genbank AN 6678787) were generated is described herein, with a similar approach being used for the other mannosidases. FIG. 3 illustrates the ORF of the M musculus α-1,2-mannosidase IA with the predicted transmembrane and catalytic domains being highlighted in bold. Based on this structure, three 5′-primers were designed (annealing positions underlined in FIG. 3) to generate the Δ65-, Δ105- and Δ187-N-terminal deletions. Using the Δ65 N-terninal deletion as an example the 5′-primer used was 5′-GGCGCGCCGACTCCTCCAAGCTGCTCAGCGGGGTCCTGTTCCAC-3′ (SEQ ID NO:54) (with the AscI restriction site highlighted in bold) in conjunction with the 3′-primer 5′-CCTTAATTAATCATTTCTCTTTGCCATCAATTTCCTTCTTCTGTTCACGG-3′ (SEQ ID NO:55) (with the PacI restriction site highlighted in bold). Both of these primers were used to amplify a 1561 bp fragment under the conditions outlined above for amplifying the full length M. musculus mannosidase 1A ORF. Furthermore, like the product obtained for the full length ORF, the truncated product was also incubated with Taq DNA polymerase, ligated into pCR2.1-TOPO (Invitrogen, Carlsbad, Calif.), transformed into TOP10 and ABI sequenced. After having amplified and confirmed the sequence of the truncated mannosidase fragment, the resulting plasmid, pCR2.1-Δ65 mMannIA, was digested with AscI and PacI in New England Biolabs buffer #4 (Beverly, Mass.) for 16 h at 37° C. In parallel, the pJN347 (FIG. 2C) was digested with the same enzymes and incubated as described above. Post-digestion, both the pJN347 (FIG. 2C) back-bone and the truncated catalytic domain were gel extracted and ligated using the Quick Ligation Kit (New England Biolabs, Beverly, Mass.), as recommended by the manufacturers, and transformed into chemically competent DH5α cells (Invitrogen, Carlsbad, Calif.). Colony PCR was used to confirm the generation of the pJN347-mouse Mannosidase IAΔ65 construct.
  • Having generated a library of truncated α-1,2-mannosidase catalytic domains in the yeast expression vector pJN347 (FIG. 2C) the remaining step in generating the targeting peptide/catalytic domain library was to clone in-frame the targeting peptide sequences (FIG. 2). Both the pJN347-mannosidase constructs (FIG. 2D) and the pCR2.1TOPO-targeting peptide constructs (FIG. 2B) such as were incubated overnight at 37° C. in New England Biolabs buffer #4 in the presence of the restriction enzymes NotI and AscI. Following digestion, both the pJN347-mannosidase back-bone and the targeting peptide regions were gel-extracted and ligated using the Quick Ligation Kit (New England Biolabs, Beverly, Mass.), as recommended by the manufacturers, and transformed into chemically competent DH5α cells (Invitrogen, Carlsbad, Calif.). Subsequently, the pJN347-targeting peptide/mannosidase constructs were ABI sequenced to confirm that the generated fusions were in-frame. The estimated size of the final targeting peptide/alpha-1,2-mannosidase library contains over 1300 constructs generated by the approach described above. FIG. 2 illustrates construction of the combinatorial DNA library.
  • Engineering a P. pastoris OCH1 Knock-out Strain with Multiple Auxotrophic Markers
  • The first step in plasmid construction involved creating a set of universal plasmids containing DNA regions of the KEX1 gene of P. pastoris (Boehm et al. Yeast 1999 May;15(7):563-72) as space holders for the 5′ and 3′ regions of the genes to be knocked out. The plasmids also contained the S. cerevisiae Ura-blaster (Alani et al. (1987) Genetics 116:541-545) as a space holder for the auxotrophic markers, and an expression cassette with a multiple cloning site for insertion of a foreign gene. A 0.9-kb fragment of the P. pastoris KEXI-5′ region was amplified by PCR using primers GGCGAGCTCGGCCTACCCGGCCAAGGCTGAGATCATTTGTCCAGCTTCA GA (SEQ ID NO:56) and GCCCACGTCGACGGATCCGTTTAAACATCGATTGGAGAGGCTGACACC GCTACTA (SEQ ID NO:57) and P. pastoris genomic DNA as a template and cloned into the SacI, SalI sites of pUC19 (New England Biolabs, Beverly, Mass.). The resulting plasmid was cut with BamHI and SalI, and a 0.8-kb fragment of the KEX1-3′ region that had been amplified using primers CGGGATCCACTAGTATTTAAATCATATGTGCGAGTGTACAACTCTTCCC ACATGG (SEQ ID NO:58) and GGACGCGTCGACGGCCTACCCGGCCGTACGAGGAATTTCTCGG ATGACTCTTTTC (SEQ ID NO:59) was cloned into the open sites creating pJN262. This plasmid was cut with BamHI and the 3.8-kb BamHI, BglII fragment of pNKY51 (Alani et al. (1987) Genetics 116:541-545) was inserted in both possible orientations resulting in plasmids pJN263 (FIG. 4A) and pJN284 (FIG. 4B).
  • An expression cassette was created with NotI and PacI as cloning sites. The GAPDH promoter of P. pastoris was amplified using primers CGGGATCCCTCGAGAGATCTTTTTTGTAGAAATGTCTTGGTGCCT (SEQ ID NO:60) and GGACATGCATGCACTAGTGCGGCCGCCACGTGATAGTTGTTCA ATTGATTGAAATAGGGACAA (SEQ ID NO:61) and plasmid pGAPZ-A (Invitrogen) as template and cloned into the BamHI, SphI sites of pUC 19 (New England Biolabs, Beverly, Mass.) (FIG. 4B). The resulting plasmid was cut with SpeI and SphI and the CYC1 transcriptional terminator region (“TT”) that had been amplified using primers CCTTGCTAGCTTAATTAACCGCGGCACGTCCGACGGCGGCCCA CGGGTCCCA (SEQ ID NO:62) and GGACATGCATGCGGATCCCTTAAGAGCCGGCAGCTTGCAAATT AAAGCCTTCGAGCGTCCC (SEQ ID NO:63) and plasmid pPICZ-A (Invitrogen) as a template was cloned into the open sites creating pJN261 (FIG. 4B).
  • A knockout plasmid for the P. pastoris OCH1 gene was created by digesting pJN263 with SalI and SpeI and a 2.9-kb DNA fragment of the OCH1-5′ region, which had been amplified using the primers GAACCACGTCGACGGCCATTGCGGCCAAAACCTTTTTTCCTATT CAAACACAAGGCATTGC (SEQ ID NO:64) and CTCCAATACTAGTCGAAGATTATCTTCTACGGTGCCTGGACTC (SEQ ID NO:65) and P. pastoris genomic DNA as a template, was cloned into the open sites (FIG. 4C). The resulting plasmid was cut with EcoRI and PmeI and a 1.0-kb DNA fragment of the OCH1-3′ region that had been generated using the primers TGGAAGGTTTAAACAAAGCTAGAGTAAAATAGATATAGCGAG ATTAGAGAATG (SEQ ID NO:66) and AAGAATTCGGCTGGAAGGCCTTGTACCTTGATGTAGTTCCCGTT TTCATC (SEQ ID NO:67) was inserted to generate pJN298 (FIG. 4C). To allow for the possibility to simultaneously use the plasmid to introduce a new gene, the BamHI expression cassette of pJN261 (FIG. 4B) was cloned into the unique BamHI site of pJN298 (FIG. 4C) to create pJN299 (FIG. 4E).
  • The P. pastoris Ura3-blaster cassette was constructed using a similar strategy as described in Lu et al. (1998) Appl. Microbiol. Biotechnol. 49:141-146. A 2.0-kb PstI, Spel fragment of P. pastoris URA3 was inserted into the PstI, XbaI sites of pUC19 (New England Biolabs, Beverly, Mass.) to create pJN306 (FIG. 4D). Then a 0.7-kb SacI, PvuII DNA fragment of the lacZ open reading frame was cloned into the Sacl, SmaI sites to yield pJN308 (FIG. 4D). Following digestion of pJN308 (FIG. 4D) with PstI, and treatment with T4 DNA polymerase, the SacI-PvuII fragment from lacZ that had been blunt-ended with T4 DNA polymerase was inserted generating pJN315 (FIG. 4D). The lacZ/URA3 cassette was released by digestion with SacI and SphI, blunt ended with T4 DNA polymerase and cloned into the backbone of pJN299 that had been digested with PmeI and AflII and blunt ended with T4 DNA polymerase. The resulting plasmid was named pJN329 (FIG. 4E).
  • A HIS4 marked expression plasmid was created by cutting pJN261 (FIG. 4F) with EcoICRI (FIG. 4F). A 2.7 kb fragment of the Pichia pastoris HIS4 gene that had been amplified using the primers GCCCAAGCCGGCCTTAAGGGATCTCCTGATGACTGACTCACTGATAATA AAAATACGG (SEQ ID NO:68) and GGGCGCGTATTTAAATACTAGTGGATCTATCGAATCTAAATGTAAGTTA AAATCTCTAA (SEQ ID NO:69) cut with NgoMIV and SwaI and then blunt-ended using T4 DNA polymerase, was then ligated into the open site. This plasmid was named pJN337 (FIG. 4F). To construct a plasmid with a multiple cloning site suitable for fusion library construction, pJN337 was cut with NotI and PacI and the two oligonucleotides GGCCGCCTGCAGATTTAAATGAATTCGGCGCGCCTTAAT (SEQ ID NO:70) and TAAGGCGCGCCGAATTCATTTAAATCTGCAGGGC (SEQ ID NO:71) that had been annealed in vitro were ligated into the open sites, creating pJN347 (FIG. 4F).
  • To create an och1 knockout strain containing multiple auxotrophic markers, 100 μg of pJN329 was digested with SfiI and used to transform P. pastoris strain JC308 (Cereghino et al. (2001) Gene 263:159-169) by electroporation. Following transformation, the URA dropout plates were incubated at room temperature for 10 days. One thousand (1000) colonies were picked and restreaked. All 000 clones were then streaked onto 2 sets of URA dropout plates. One set was incubated at room temperature, whereas the second set was incubated at 37° C. The clones that were unable to grow at 37° C., but grew at room temperature, were subjected to colony PCR to test for the correct OCH1 knockout. One clone that showed the expected PCR signal (about 4.5 kb) was designated YJN153.
  • EXAMPLE 5 Characterization of the Combinatorial DNA Library
  • Positive transformants screened by colony PCR confirming integration of the mannosidase construct into the P. pastoris genome were subsequently grown at room temperature in 50 ml BMGY buffered methanol-complex medium consisting of 1% yeast extract, 2% peptone, 100 mM potassium phosphate buffer, pH 6.0, 1.34% yeast nitrogen base, 4×10−5% biotin, and 1% glycerol as a growth medium) until OD600nm 2-6 at which point they were washed with 10 ml BMMY (buffered methanol-complex medium consisting of I % yeast extract, 2% peptone, 100 mM potassium phosphate buffer, pH 6.0, 1.34% yeast nitrogen base, 4×10−5% biotin, and 1.5% methanol as a growth medium) media prior to induction of the reporter protein for 24 hours at room temperature in 5 ml BMMY. Consequently, the reporter protein was isolated and analyzed as described in Example 3 to characterize its glycan structure. Using the targeting peptides in Table 6, mannosidase catalytic domains localized to either the ER or the Golgi showed significant level of trimming of a glycan predominantly containing Man8GlcNAc2 to a glycan predominantly containing Man5GlcNAc2. This is evident when the glycan structure of the reporter glycoprotein is compared between that of P. pastoris och1 knock-out in FIGS. 5C and 6C and the same strain transformed with M. musculus mannosidase constructs as shown in FIGS. 5D, 5E, 6D-6F. FIGS. 5 and 6 show expression of constructs generated from the combinatorial DNA library which show significant mannosidase activity in P. pastoris. Expression of pGC5 (Saccharomyces MNS1 (m)/mouse mannosidase IB Δ99) (FIGS. 5D and 6E) produced a protein which has approximately 30% of all glycans trimmed to Man5GlcNAc2, while expression of pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187) (FIG. 6F) produced approximately 50% Man5GlcNAc2 and expression of pBC18-5 (Saccharomyces VAN1 (s)/C. elegans mannosidase IB Δ80) (FIG. 5E) produced 70% Man5GlcNAc2.
  • EXAMPLE 6 Trimming In Vivo by αt-1,2-mannosidase
  • To ensure that the novel engineered strains of Example 4 in fact produced the desired Man5GlcNAc2 structure in vivo, cell supernatants were tested for mannosidase activity (see FIGS. 7-9). For each construct/host strain described below, HPLC was performed at 30° C. with a 4.0 mm×250 mm column of Altech (Avondale, Pa., USA) Econosil-NH2 resin (5 μm) at a flow rate of 1.0 ml/min for 40 min. In FIGS. 7 and 8, degradation of the standard Man9GlcNAc2 [b] was shown to occur resulting in a peak which correlates to Man8GlcNAc2. In FIG. 7, the Man9GlcNAc2[b] standard eluted at 24.61 min and Man5GlcNAc2 [a] eluted at 18.59 min. In FIG. 8, Man9GlcNAc2 eluted at 21.37 min and Man5GlcNAc2 at 15.67 min. In FIG. 9, the standard Man8GlcNAc2[b] was shown to elute at 20.88 min.
  • P. pastoris cells comprising plasmid pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187) were grown at 30° C. in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for mannosidase assays and the remainder was used for the recovery of secreted soluble K3. A single purification step using CM-sepharose chromatography and an elution gradient of 25 mM NaAc, pH5.0 to 25 mM NaAc, pH5.0, 1M NaCl, resulted in a 95% pure K3 eluting between 300-500 mM NaCl. N-glycan analysis of the K3 derived glycans is shown in FIG. 6F. The earlier removed aliquot of the supernatant was further tested for the presence of secreted mannosidase activity. A commercially available standard of 2-aminobenzamide-labeled N-linked-type oligomannose 9 (Man9-2-AB) (Glyko, Novato, Calif.) was added to: BMMY (FIG. 7A), the supernatant from the above aliquot (FIG. 7B), and BMMY containing 10 ng of 75 mU/mL of α-1,2-mannosidase from Trichoderma reesei (obtained from Contreras et al., WO 02/00856 A2) (FIG. 7C). After incubation for 24 hours at room temperature, samples were analyzed by amino silica HPLC to determine the extent of mannosidase trimming.
  • P. pastoris cells comprising plasmid pGC5 (Saccharomyces MNS1 (m)/mouse mannosidase IB Δ99) were similarly grown and assayed. Cells were grown at room temperature in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for mannosidase assays and the remainder was used for the recovery of secreted soluble K3. A single purification step using CM-sepharose chromatography and an elution gradient of 25 mM NaAc, pH5.0 to 25 mM NaAc, pH5.0, 1 M NaCl, resulted in a 95% pure K3 eluting between 300-500 mM NaCl. N-glycan analysis of the K3 derived glycans is shown in FIG. 5D. The earlier removed aliquot of the supernatant was further tested for the presence of secreted mannosidase activity as shown in FIG. 8B. A commercially available standard of Man9-2-AB (Glyko, Novato, Calif.) were added to:BMMY (FIG. 8A), supernatant from the above aliquot (FIG. 8B), and BMMY containing 10 ng of 75 mU/mL of α-1,2-mannosidase from Trichoderma reesei (obtained from Contreras et al., WO 02/00856 A2) (FIG. 8C). After incubation for 24 hours at room temperature, samples were analyzed by amino silica HPLC to determine the extent of mannosidase trimming.
  • Man9-2-AB was used as a substrate and it is evident that after 24 hours of incubation, mannosidase activity was virtually absent in the supernatant of the pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187) strain digest (FIG. 7B) and pGC5 (Saccharomyces MNS1 (m)/mouse mannosidase IB Δ99) strain digest (FIG. 8B) whereas the positive control (purified α-1,2-mannosidase from T. reesei obtained from Contreras) leads to complete conversion of Man9GlcNAc2 to Man5GlcNAc2 under the same conditions, as shown in FIGS. 7C and 8C. This is conclusive data showing in vivo mannosidase trimming in P. pastoris pGC5 strain; and pFB8 strain, which is distinctly different from what has been reported to date (Contreras et al., WO 02/00856 A2) .
  • FIG. 9 further substantiates localization and activity of the mannosidase enzyme. P. pastoris comprising pBC18-5 (Saccharomyces VAN1 (s)/C. elegans mannosidase IB Δ80) was grown at room temperature in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for mannosidase assays and the remainder was used for the recovery of secreted soluble K3. A single purification step using CM-sepharose chromatography and an elution gradient 25 mM NaAc, pH5.0 to 25 mM NaAc, pH5.0, 1M NaCl, resulted in a 95% pure K3 eluting between 300-500 mM NaCl. N-glycan analysis of the K3 derived glycans is shown in FIG. 5E. The earlier removed aliquot of the supernatant was further tested for the presence of secreted mannosidase activity as shown in FIG. 9B. A commercially available standard of Man8-2-AB (Glyko, Novato, Calif.) was added to:BMMY (FIG. 9A), supernatant from the above aliquot pBC18-5 (Saccharomyces VAN1 (s)/C. elegans mannosidase IB Δ80) (FIG. 9B), and BMMY containing media from a different fusion construct pDD28-3 (Saccharomyces MNN10 (m) (from SwissProt 50108)/H. sapiens mannosidase IB Δ99) (FIG. 9C). After incubation for 24 hours at room temperature,. samples were analyzed by amino silica HPLC to determine the extent of mannosidase trimming. FIG. 9B demonstrates intracellular mannosidase activity in comparison to a fusion construct pDD28-3 (Saccharomyces MNN10 (m) H. sapiens mannosidase IB Δ99) exhibiting a negative result (FIG. 9C).
  • EXAMPLE 7 pH Optimum Assay of Engineered α-1,2-Mannosidase
  • P. pastoris cells comprising plasmid pBB27-2 (Saccharomyces MANN10 (s) (from SwissProt 50108)/C. elegans mannosidase IB Δ31) were grown at room temperature in BMGY to an OD600 of about 17. About 80 μL of these cells were inoculated into 600 μL BMGY and were grown overnight. Subsequently, cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant (pH 6.43). The supernatant was removed for mannosidase pH optimum assays. Fluorescence-labeled Man8GlcNAc2 (0.5 μg) was added to 20 μL of supernatant adjusted to various pH (FIG. 11) and incubated for 8 hours at room temperature. Following incubation the sample was analyzed by HPLC using an Econosil NH2 4.6×250 mm, 5 micron bead, amino-bound silica column (Altech, Avondale, Pa.). The flow rate was 1.0 ml/min for 40 min and the column was maintained to 30° C. After eluting isocratically (68% A:32% B) for 3 min, a linear solvent gradient (68% A:32% B to 40% A:60% B) was employed over 27 min to elute the glycans (18). Solvent A (acetonitrile) and solvent B (ammonium formate, 50 mM, pH 4.5. The column was equilibrated with solvent (68% A:32% B) for 20 min between runs.
  • EXAMPLE 8 Engineering of P. pastoris to Produce N-glycans with the Structure GlcNAcMan5GlcNAc2
  • GlcNAc Transferase I activity is required for the maturation of complex and hybrid N-glycans (U.S. Pat. No. 5,834,251). Man5GlcNAc2 may only be trimmed by mannosidase II, a necessary step in the formation of human glycoforms, after the addition of N-acetylglucosamine to the terminal α-1,3 mannose residue of the trimannose stem by GlcNAc Transferase I (Schachter, 1991 Glycobiology 1(5):453-461). Accordingly, a combinatorial DNA library was prepared including DNA fragments encoding suitably targeted catalytic domains of GlcNAc Transferase I genes from C. elegans and Homo sapiens; and localization sequences from GLS, MNS, SEC, MNN9, VAN1, ANP1, HOC1, MNN10, MNN11, MNT1, KTR1, KTR2, MNN2, MNN5, YUR1, MNN1, and MNN6 from S. cerevisiae and P. pastoris putative α-1,2-mannosyltransferases based on the homology from S. cerevisiae: D2, D9 and J3, which are KTR homologs. Table 10 includes but does not limit targeting peptide sequences such as SEC and OCH1, from P. pastoris and K. lactis GnTI, (See Table 6 and Table 10)
    TABLE 10
    A representative combinatorial library of targeting peptide sequences/
    catalytic domain for UDP-N-Acetylglucosaminyl Transferase I (GnTI)
    Targeting peptide
    OCHI(s) OCHI(m) OCHI(l) MNN9(s) MNN9(m)
    Catalytic Human, GnTI, Δ38 PB105 PB106 PB107 PB104 N/A
    Domain Human, GnTI, Δ86 NB12 NB13 NB14 NB15 NB
    C. elegans, GnTI, Δ88 OA12 OA13 OA14 OA15 OA16
    C. elegans, GnTI, Δ35 PA12 PA13 PA14 PA15 PA16
    C. elegans, GnTI, Δ63 PB12 PB13 PB14 PB15 PB16
    X. leavis, GnTI, Δ33 QA12 QA13 QA14 QA15 QA16
    X. leavis, GnTI, Δ103 QB12 QB13 QB14 QB15 QB16
  • Targeting peptide sequences were selected from OCH1 in P. pastoris (long, medium and short) (see Example 4) and MNN9 (SwissProt P39107) in S. cerevisiae (short and medium). Catalytic domains were selected from human GnTI with a 38 and 86 amino acid N-terminal deletion, C. elegans (gly-12) GnTI with a 35 and 63 amino acid deletion as well as C. elegans (gly-14) GnTI with a 88 amino acid N-terminal deletion and X. leavis GnTI with a 33 and 103 amino acid N-terminal deletion, respectively.
  • A portion of the gene encoding human N-acetylglucosaminyl Transferase I (MGATI, Accession# NM002406), lacking the first 154 bp, was amplified by PCR using oligonucleotides 5′-TGGCAGGCGCGCCTCAGTCAGCGCTCTCG-3′ (SEQ ID NO:72) and 5′-AGGTTAATTA AGTGCTAATTCCAGCTAGG-3′ (SEQ ID NO:73) and vector pHG4.5 (ATCC# 79003) as template. The resulting PCR product was cloned into pCR2.1-TOPO and the correct sequence was confirmed. Following digestion with AscI and PacI the truncated GnTI was inserted into plasmid pJN346 to create pNA. After digestion of pJN271 with NotI and AscI, the 120 bp insert was ligated into pNA to generate an in-frame fusion of the MNN9 transmembrane domain with the GnTI, creating pNA 15.
  • The host organism is a strain of P. pastoris that is deficient in hypermannosylation (e.g. an och1 mutant), provides the substrate UDP-GlcNAc in the Golgi and/or ER (i.e., contains a functional UDP-GlcNAc transporter), and provides N-glycans of the structure Man5GlcNAc2 in the Golgi and/or ER (e.g. P. pastoris pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187) from above). First, P. pastoris pFB8 was transformed with pPB103 containing the Kluyveromyces lactis MAN2-2 gene (Genbank AN AF 106080) (encoding UDP-GlcNAc transporter) cloned into BamHI and BglII site of pBLADE-SX plasmid (Cereghino et al. (2001) Gene 263:159-169). Then the aforementioned combinatorial DNA library encoding a combination of exogenous or endogenous GnTI/localization genes was transformed and colonies were selected and analyzed for the presence of the GnTI construct by colony PCR. Our transformation and integration efficiency was generally above 80% and PCR screening can be omitted once robust transformation parameters have been established.
  • Protein Purification
  • K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot. The robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin. Another screening method may be performed using a specific terminal GlcNAc binding antibody, or a lectin such as the GSII lectin from Griffonia simplificolia, which binds terminal GlcNAc (EY Laboratories, San Mateo, Calif.). These screens can be automated by using lectins or antibodies that have been modified with fluorescent labels such as FITC or analyzed by MALDI-TOF.
  • Secreted K3 can be purified by Ni-affinity chromatography, quantified and equal amounts of protein can be bound to a high protein binding 96-well plate. After blocking with BSA, plates can be probed with a GSII-FACS lectin and screened for maximum fluorescent response. A preferred method of detecting the above glycosylated proteins involves the screening by MALDI-TOF mass spectrometry following the affinity purification of secreted K3 from the supernatant of 96-well cultured transformants. Transformed colonies were picked and grown to an OD600 of 10 in a 2 ml, 96-well plate in BMGY at 30° C. Cells were harvested by centrifugation, washed in BMMY and resuspended in 250 ul of BMMY. Following 24 hours of induction, cells were removed by centrifugation, the supernatant was recovered and K3 was purified from the supernatant by Ni affinity chromatography. The N-glycans were released and analyzed by MALDI-TOF delayed extraction mass spectrometry as described herein.
  • In summary, the methods of the invention yield strains of P. pastoris that produce GlcNAcMan5GlcNAc2 in high yield, as shown in FIG. 10B. At least 60% of the N-glycans are GlcNAcMan5GlcNAc2. To date, no report exists that describes the formation of GlcNAcMan5GlcNAc2 on secreted soluble glycoproteins in any yeast. Results presented herein show that addition of the UDP-GlcNAc transporter along with GnTI activity produces a predominant GlcNAcMan5GlcNAc2 structure, which is confirmed by the peak at 1457 (m/z) (FIG. 10B).
  • Construction of Strain PBP-3:
  • The P. pastoris strain expressing K3, (Δoch1, arg-, ade-, his-) was transformed successively with the following vectors. First, pFB8 (Saccharomyces SEC12 (m)/mouse mannosidase IA Δ187) was transformed in the P. pastoris strain by electroporation. Second, pPB103 containing Kluyveromyces lactis MNN2-2 gene (Genbank AN AF 106080) (encoding UDP-GlcNAc transporter) cloned into pBLADE-SX plasmid (Cereghino et al. (2001) Gene 263:159-169) digested with BamHI and BgIII enzymes was transformed in the P. pastoris strain. Third, pPB104 containing Saccharomyces MNN9(s)/human GnTI Δ38 encoding gene cloned as NotI-PacI fragment into pJN336 was transformed into the P. pastoris strain.
  • EXAMPLE 9 Engineering K. lactis Cells to Produce N-glycans with the Structure Man5GlcNAC2
  • Identification and Disruption of the K. lactis OCH1 Gene
  • The OCH1 gene of the budding yeast S. cerevisiae encodes a 1,6-mannosyltransferase that is responsible for the first Golgi localized mannose addition to the Man8GlcNAc2 N-glycan structure on secreted proteins (Nakanishi-Shindo et al. (1993) J. Biol. Chem.; 268(35):26338-45). This mannose transfer is generally recognized as the key initial step in the fungal specific polymannosylation of N-glycan structures (Nakanishi-Shindo et al. (1993) J. Biol. Chem. 268(35):26338-26345; Nakayama et al. (1992) EMBO J. 11(7):2511-19; Morin-Ganet et al (2000) Traffic 1(1):56-68). Deletion of this gene in S. cerevisiae results in a significantly shorter N-glycan structure that does not include this typical polymannosylation or a growth defect at elevated temperatures (Nakayama et al. (1992) EMBO J. 11 (7):2511-19).
  • The Och1p sequence from S. cerevisiae was aligned with known homologs from Candida albicans (Genbank accession # AAL49987), and P. pastoris along with the Hoc1 proteins of S. cerevisiae (Neiman et al (1997) Genetics 145(3):637-45 and K. lactis (PENDANT EST database) which are related but distinct mannosyltransferases. Regions of high homology that were in common among Och1p homologs but distinct from the Hoc1p homologs were used to design pairs of degenerate primers that were directed against genomic DNA from the K. lactis strain MG1/2 (Bianchi et al (1987) Current Genetics 12:185-192). PCR amplification with primers RCD33 (CCAGAAGAATTCAATTYTGYCARTGG) (SEQ ID NO:74) and RCD34 (CAGTGAAAATACCTGGNCCNGTCCA) (SEQ ID NO:75) resulted in a 302 bp product that was cloned and sequenced and the predicted translation was shown to have a high degree of homology to Och1 proteins (>55% to S. cerevisiae Och1p).
  • The 302 bp PCR product was used to probe a Southern blot of genomic DNA from K. lactis strain (MG1/2) with high stringency (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989). Hybridization was observed in a pattern consistent with a single gene indicating that this 302 bp segment corresponds to a portion of the K. lactis genome and K. lactis (KiOCH1) contains a single copy of the gene. To clone the entire KlOCH1 gene, the Southern blot was used to map the genomic locus. Accordingly, a 5.2 kb BamHI/PstI fragment was cloned by digesting genomic DNA and ligating those fragments in the range of 5.2 kb into pUC 19 (New England Biolabs, Beverly, Mass.) to create a K. lactis subgenomic library. This subgenomic library was transformed into E. coli and several hundred clones were tested by colony PCR using RCD 33/34. The 5.2 kb clone containing the predicted KiOCH1 gene was sequenced and an open reading frame of 1362 bp encoding a predicted protein that is 46.5% identical to the S. cerevisiae OCH1 gene. The 5.2 kb sequence was used to make primers for construction of an och1::KANR deletion allele using a PCR overlap method (Davidson et al. (2002) Microbiol. 148(Pt 8):2607-15). This deletion allele was transformed into two K. lactis strains and G418 resistant colonies selected. These colonies were screened by both PCR and for temperature sensitivity to obtain a strain deleted for the OCH1 ORF. The results of the experiment show strains which reveal a mutant PCR pattern, which were characterized by analysis of growth at various temperatures and N-glycan carbohydrate analysis of secreted and cell wall proteins following PNGase digestion. The och1 mutation conferred a temperature sensitivity which allowed strains to grow at 30° C. but not at 35° C. FIG. 12A shows a MALDI-TOF analysis of a wild type K. lactis strain producing N-glycans of Man8GlcNAc2[c] and higher.
  • Identification, Cloning, and Disruption of the K. lactis MNN1 Gene
  • S. cerevisiae MANN1 is the structural gene for the Golgi α-1,3-mannosyltransferase. The product of MNN1 is a 762-amino acid type II membrane protein (Yip et al. (1994) Proc Natl Acad Sci USA. 91(7):2723-7). Both N-linked and O-linked oligosaccharides isolated from mnn1 mutants lack α-1,3-mannose linkages (Raschke et al. (1973) J. Biol Chem. 248(13):4660-66).
  • The Mnn1p sequence from S. cerevisiae was used to search the K. lactis translated genomic sequences (PEDANT). One 405 bp DNA sequence encoding a putative protein fragment of significant similarity to Mnn1p was identified. An internal segment of this sequence was subsequently PCR amplified with primers KMN1 (TGCCATCTTTTAGGTCCAGGCCCGTTC) (SEQ ID NO:76) and KMN2 (GATCCCACGACGCATCGTATTTCTTTC), (SEQ ID NO:77) and used to probe a Southern blot of genomic DNA from K. lactis strain (MG 1/2). Based on the Southern hybridization data a 4.2 Kb BamHI-PstI fragment was cloned by generating a size-selected library as described herein. A single clone containing the K. lactis MNN1 gene was identified by whole colony PCR using primers KMN1 and KMN2 and sequenced. Within this clone a 2241 bp ORF was identified encoding a predicted protein that was 34% identical to the S. cerevisiae MNN1 gene. Primers were designed for construction of a mnn1::NATR deletion allele using the PCR overlap method (Davidson et al. (2002) Microbiol. 148(Pt 8):2607- 15).
  • This disruption allele was transformed into a strain of K. lactis by electroporation and nourseothricin resistant transformants were selected and PCR amplified for homologous insertion of the disruption allele. Strains that reveal a mutant PCR pattern may be subjected to N-glycan carbohydrate analysis of a known reporter gene.
  • FIG. 12B depicts the N-glycans from the K. lactis och1 mnn1 deletion strain observed following PNGase digestion the MALDI-TOF as described herein. The predominant peak at 1908 (m/z) indicated as [d] is consistent with the mass of Man9GlcNAc2.
  • Additional methods and reagents which can be used in the methods for modifying the glycosylation are described in the literature, such as U.S. Pat. No. 5,955,422, U.S. Pat. No. 4,775,622, U.S. Pat. No. 6,017,743, U.S. Pat. No. 4,925,796, U.S. Pat. No. 5,766,910, U.S. Pat. No. 5,834,251, U.S. Pat. No. 5,910,570, U.S. Pat. No. 5,849,904, U.S. Pat. No. 5,955,347, U.S. Pat. No. 5,962,294, U.S. Pat. No. 5,135,854, U.S. Pat. No. 4,935,349, U.S. Pat. No. 5,707,828, and U.S. Pat. No. 5,047,335. Appropriate yeast expression systems can be obtained from sources such as the American Type Culture Collection, Rockville, Md. Vectors are commercially available from a variety of sources.
  • EXAMPLE 10 Identification, Cloning and Deletion of the ALG3 Gene in P. pastoris and K. lactis
  • Degenerate primers were generated based on an alignment of Alg3 protein sequences from S. cerevisiae, H. sapiens, and D. melanogaster and were used to amplify an 83 bp product from P. pastoris genomic DNA:
    (SEQ ID NO:78)
    5′-GGTGTTTTGTTTTCTAGATCTTTGCAYTAYCARTT-3′
    and
    (SEQ ID NO:79)
    5′-AGAATTTGGTGGGTAAGAATTCCARCACCAYTCRTG-3′.
  • The resulting PCR product was cloned into the pCR2.1 vector (Invitrogen, Carlsbad, Calif.) and seqence analysis revealed homology to known ALG3/RHK1/NOT56 homologs (Genbank NC001134.2, AF309689, NC003424.1). Subsequently, 1929 bp upstream and 2738 bp downstream of the initial PCR product were amplified from a P. pastoris genomic DNA library (Boehm (1999) Yeast 15(7):563-72) using the internal oligonucleotides 5′- CCTAAGCTGGTATGCGTTCTCTTTGCCATATC-3′ (SEQ ID NO:80) and 5′-GCGGCATAAACAATAATAGATGCTATAAAG-3′ (SEQ ID NO:81) along with T3 (5′-AATTAACCCTCACTAAAGGG-3′) (SEQ ID NO:49) and T7 (5′-GTAA TACGACTCACTATAGGGC-3′) (SEQ ID NO:48) (Integrated DNA Technologies, Coralville, Iowa) in the backbone of the library bearing plasmid lambda ZAP II (Stratagene, La Jolla, Calif.). The resulting fragments were cloned into the pCR2.1-TOPO vector (Invitrogen) and sequenced. From this sequence, a 1395 bp ORF was identified that encodes a protein with 35% identity and 53% similarity to the S. cerevisiae ALG3 gene (using BLAST programs). The gene was named PpALG3.
  • The sequence of PpALG3was used to create a set of primers to generate a deletion construct of the PpALG3 gene by PCR overlap (Davidson et al (2002) Microbiol. 148(Pt 8):2607-15). Primers below were used to amplify 1 kb regions 5′ and 3′ of the PpALG3 ORF and the KANR gene, respectively:
    RCD142 (5′-CCACATCATCCGTGCTACATATAG-3′), (SEQ ID NO:82)
    RCD144 (5′-ACGAGGCAAGCTAAACAGATCTCGAAGTATCGAGGGTTATCCAG-3′), (SEQ ID NO:83)
    RCD145 (5′-CCATCCAGTGTCGAAAACGAGCCAATGGTTCATGTCTATAAATC-3′), (SEQ ID NO:84)
    RCD147 (5′-AGCCTCAGCGCCAACAAGCGATGG-3′), (SEQ ID NO:85)
    RCD143 (5′-CTGGATAACCCTCGATACTTCGAGATCTGTTTAGCTTGCCTCGT-3′), (SEQ ID NO:86)
    and
    RCD146 (5′-GATTTATAGACATGAACCATTGGCTCGTTTTCGACACTGGATGG-3′). (SEQ ID NO:87)
  • Subsequently, primers RCD142 and RCD147 were used to overlap the three resulting PCR products into a single 3.6 kb alg3::KANR deletion allele. Identification, cloning and deletion of the ALG3 gene in K. lactis.
  • The ALG3p sequences from S. cerevisiae, Drosophila melanogaster, Homo sapiens etc were aligned with K. lactis sequences (PENDANT EST database). Regions of high homology that were in common homologs but distinct in exact sequence from the homologs were used to create pairs of degenerate primers that were directed against genomic DNA from the K. lactis strain MG 1/2 (Bianchi et al, 1987). In the case of ALG3, PCR amplification with primers KAL-1 (5′-ATCCTTTACCGATGCTGTAT-3′) (SEQ ID NO:88) and KAL-2 (5′-ATAACAGTATGTGTTACACGCGTGTAG-3′) (SEQ ID NO:89) resulted in a product that was cloned and sequenced and the predicted translation was shown to have a high degree of homology to Alg3p proteins (>50% to S. cerevisiae Alg3p).
  • The PCR product was used to probe a Southern blot of genomic DNA from K. lactis strain (MG1/2) with high stringency (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989). Hybridization was observed in a pattern consistent with a single gene. This Southern blot was used to map the genomic loci. Genomic fragments were cloned by digesting genomic DNA and ligating those fragments in the appropriate size-range into pUC 19 to create a K. lactis subgenomic library. This subgenomic library was transformed into E. coli and several hundred clones were tested by colony PCR, using primers KAL-1 and KAL-2. The clones containing the predicted KiALG3 and KlALG61 genes were sequenced and open reading frames identified.
  • Primers for construction of an alg3::NATR deletion allele, using a PCR overlap method (Davidson et al. (2002) Microbiol. 148(Pt 8):2607-15), were designed and the resulting deletion allele was transformed into two K. lactis strains and NAT-resistant colonies selected. These colonies were screened by PCR and transformants were obtained in which the ALG3 ORF was replaced with the och1::NATR mutant allele.
  • EXAMPLE 11 Generation of an alg3 och1 Mutant Strain Expressing an α-1,2-Mannosidase, GnT1 and GnTII for Production of a Human-Like Glycoprotein
  • A P. pastoris alg3::KANR deletion construct was generated as described in Example 10. Approximately 5 μg of the resulting PCR product was transformed into strain PBP-3 (see Example 3), and colonies were selected on YPD medium containing 200 μg/ml G418. One strain out of 20 screened by PCR was confirmed to contain the correct integration of the alg3::KANR mutant allele and lack the wild-type allele. This strain was named RDP27 (FIG. 36).
  • A library of GnTII constructs was then generated, which was comprised of in-frame fusions of the leader library with the catalytic domains of GnTII genes from human and rat sources (WO 02/00879). This library was created in a P. pastoris integration vector containing the NSTR gene conferring resistance to the drug nourseothricin. The library plasmids were linearized with EcoRI, transformed into strain RDP27 by electroporation, and the resulting strains were screened by analysis of the released glycans from purified K3. A P. pastoris strain expressing the rat GnTII fused in-frame to the S. cerevisiae MNN9 (s) construct was named PBP6-5 (FIG. 36).
  • Generation of GnTII Expression Constructs
  • The construction of a GnTI expression vector (pNA15) containing a human GnTI gene fused with the N-terminal part of S. cerevisiae MNN9 gene is described in Choi et al. (2003) Proc Natl Acad Sci U S A. 100(9):5022-27. In a similar fashion, the rat GnTII gene was cloned. The rat GnTII gene (GenBank accession number U21662) was PCR amplified using Takara EX Taq™ polymerase (Panvera) from rat liver cDNA library (Clontech) with RAT1 (5′-TTCCTCACTGCAGTCTTCTATAACT-3′) (SEQ ID NO:90) and RAT2 (5′-TGGAGACCATGAGGTTCCGCATCTAC-3′) (SEQ ID NO:91) primers. The PCR product was then cloned into pCR2.1-TOPO vector (Invitrogen) and sequenced. Using this vector as a template, the AscI-Pacl fragment of GnTII, encoding amino-acids 88-443, was amplified with Pfu Turbo polymerase (Stratagene) and primers, RAT44 (5′-TTGGCGCGCCTCCCTAGTGTACCAGTTGAACTTTG-3′) (SEQ ID NO:92) and RAT11 (5′-GATTAATTAACTCACTGCAGTCTTCTATAACT-3′) (SEQ ID NO:93) respectively, introduced AscI and Pacd restriction sites are underlined). Following confirmation by sequencing, the catalytic domain of rat GnTIl was than cloned downstream of the PMA1 promoter as a AscI-Pacl fragment in pBP 124. In the final step, the gene fragment encoding the S. cerevisiae Mnn2 localization signal was cloned from pJN281 as a NotI-AscI fragment to generate an in-frame fusion with the catalytic domain of GnTII, to generate plasmid pTC53.
  • EXAMPLE 12 Cloning and Expression Of GnTIII To Produce Bisecting GlcNAcs Which Boost Antibody Functionality
  • The addition of an N-acetylglucosamine to the GlcNAc2Man3GlcNAc2 structure by N-acetylglucosaminyltransferases III yields a so-called bisected N-glycan (see FIG. 15). This structure has been implicated in greater antibody-dependent cellular cytotoxicity (ADCC) (Umana et al. (1999) Nat. Biotechnol. 17(2):176-80).
  • A host cell such as a yeast strain capable of producing glycoproteins with bisected N-glycans is engineered according to the invention, by introducing into the host cell a GnTIII activity. Preferably, the host cell is transformed with a nucleic acid that encodes GnTIII (e.g., a mammalian such as the murine GnTIII shown in FIG. 24) or a domain thereof having enzymatic activity, optionally fused to a heterologous cell signal targeting peptide (e.g., using the libraries and associated methods of the invention.)
  • IgGs consist of two heavy-chains (VH, C H1, C H2 and C H3 in FIG. 22), interconnected in the hinge region through three disulfide bridges, and two light chains (VL, CL in FIG. 22). The light chains (domains VL and CL) are linked by another disulfide bridge to the CHI portion of the heavy chain and together with the C H1 and VH fragment make up the so-called Fab region. Antigens bind to the terminal portion of the Fab region. The Fc region of IgGs consists of the C H3, the C H2 and the hinge region and is responsible for the exertion of so-called effector functions (see below).
  • The primary function of antibodies is binding to an antigen. However, unless binding to the antigen directly inactivates the antigen (such as in the case of bacterial toxins), mere binding is meaningless unless so-called effector-functions are triggered. Antibodies of the IgG subclass exert two major effector-functions: the activation of the complement system and induction of phagocytosis. The complement system consists of a complex group of serum proteins involved in controlling inflammatory events, in the activation of phagocytes and in the lytical destruction of cell membranes. Complement activation starts with binding of the C1 complex to the Fc portion of two IgGs in close proximity. C1 consists of one molecule, C1q, and two molecules, C1r and C1s. Phagocytosis is initiated through an interaction between the IgG's Fc fragment and Fc-gamma-receptors (FcγRI, II and III in FIG. 22). Fc receptors are primarily expressed on the surface of effector cells of the immune system, in particular macrophages, monocytes, myeloid cells and dendritic cells.
  • The C H2 portion harbors a conserved N-glycosylation site at asparagine 297 (Asn297). The Asn297 N-glycans are highly heterogeneous and are known to affect Fc receptor binding and complement activation. Only a minority (i.e., about 15-20%) of IgGs bears a disialylated, and 3-10% have a monosialylated N-glycan (reviewed in Jefferis (2001) Biopharm. 14:19-26). Interestingly, the minimal N-glycan structure shown to be necessary for fully functional antibodies capable of complement activation and Fc receptor binding is a pentasacharide with terminal N-acetylglucosamine residues (GlcNAc2Man3) (reviewed in Jefferis, R., Glycosylation of human IgG Antibodies. BioPharm, 2001). Antibodies with less than a GlcNAc2Man3 N-glycan or no N-glycosylation at Asn297 might still be able to bind an antigen but most likely will not activate the crucial downstream events such as phagocytosis and complement activation. In addition, antibodies with fungal-type N-glycans attached to Asn297 will in all likelihood solicit an immune-response in a mammalian organism which will render that antibody useless as a therapeutic glycoprotein.
  • Cloning And Expression Of GnTIII
  • The DNA fragment encoding part of the mouse GnTIII protein lacking the TM domain is PCR amplified from murine (or other mammalian) genomic DNA using forward (5′-TCCTGGCGCGCCTTCCCGAGAGAACTGGCCTCCCTC-3′) (SEQ ID NO:94) and reversed (5′-AATTAATTAACCCTAGCCCTCCGCTGTATCCAACTTG-3′) (SEQ ID NO:95) primers. Those primers include AscI and Pacl restriction sites that may be used for cloning into the vector suitable for the fusion with leader library.
  • The nucleic acid (SEQ ID NO:45) and amino acid (SEQ ID NO:46) sequences of murine GnTIII are shown in FIG. 24.
  • Cloning of Immunoglobulin-Encoding Sequences
  • Protocols for the cloning of the variable regions of antibodies, including primer sequences, have been published previously. Sources of antibodies and encoding genes can be, among others, in vitro immunized human B cells (see, e.g., Borreback et al. (1988) Proc. Natl. Acad. Sci. USA 85:3995-3999), peripheral blood lymphocytes or single human B cells (see, e.g., Lagerkvist et al. (1995) Biotechniques 18:862-869; and Terness et al. (1997) Hum. Immunol. 56:17-27) and transgenic mice containing human immunoglobulin loci, allowing the creation of hybridoma cell-lines.
  • Using standard recombinant DNA techniques, antibody-encoding nucleic acid sequences can be cloned. Sources for the genetic information encoding immunoglobulins of interest are typically total RNA preparations from cells of interest, such as blood lymphocytes or hybridoma cell lines. For example, by employing a PCR based protocol with specific primers, variable regions can be cloned via reverse transcription initiated from a sequence-specific primer hybridizing to the IgG C H1 domain site and a second primer encoding amino acids 111-118 of the murine kappaconstant region. The VH and VK encodinWDNAs can then be amplified as previously published (see, e.g., Graziano et al. (1995) J. Immunol. 155(10):4996-5002; Welschofetal. (1995) J. Immunol. Methods 179:203-214; and Orlandi et al. (1988) Proc. Natl. Acad. Sci. USA 86:3833). Cloning procedures for whole immunoglobulins (heavy and light chains) have also been published (see, e.g., Buckel et al. (1987) Gene 51:13-19; Recinos et al. (1994) Gene 149: 385-386; Recinos et al. (1995) Gene 158:311-12). Additional protocols for the cloning and generation of antibody fragment and antibody expression constructs have been described in Antibody Engineering, Kontermann and Dübel (2001), Eds., Springer Verlag: Berlin Heidelberg New York.
  • Fungal expression plasmids encoding heavy and light chain of immunoglobulins have been described (see, e.g., Abdel-Salam et al. (2001) Appl. Microbiol. Biotechnol. 56:157-164; and Ogunjimi et al. (1999) Biotechnology Letters 21:561-567). One can thus generate expression plasmids harboring the constant regions of immunoglobulins. To facilitate the cloning of variable regions into these expression vectors, suitable restriction sites can be placed in close proximity to the termini of the variable regions. The constant regions can be constructed in such a way that the variable regions can be easily in-frame fused to them by a simple restriction-digest/ligation experiment. FIG. 23 shows a schematic overview of such an expression construct, designed in a very modular way, allowing easy exchange of promoters, transcriptional terminators, integration targeting domains and even selection markers.
  • As shown in FIG. 23, VL as well as VH domains of choice can be easily cloned in-frame with CL and the CH regions, respectively. Initial integration is targeted to the P. pastoris AOX locus (or homologous locus in another fungal cell) and the methanol-inducible AOX promoter will drive expression. Alternatively, any other desired constitutive or inducible promoter cassette may be used. Thus, if desired, the 5′ AOX and 3′ AOX regions as well as transcriptional terminator (TT) fragments can be easily replaced with different TT, promoter and integration targeting domains to optimize expression. Initially the alpha-factor secretion signal with the standard KEX protease site is employed to facilitate secretion of heavy and light chains. The properties of the expression vector may be further refined using standard techniques.
  • An Ig expression vector such as the one described above is introduced into a host cell of the invention that expresses GnTIII, preferably in the Golgi apparatus of the host cell. The Ig molecules expressed in such a host cell comprise N-glycans having bisecting GlcNAcs.
  • EXAMPLE 13 Generation of Yeast Strain YSH-1 (Δoch1, α1,2-mannosidase, GnTI)
  • The previously reported P. pastoris strain BK64 (Choi et al. (2003) Proc Natl Acad Sci USA. 100(9):5022-7), a triple auxotroph (ADE, ARG, HIS) possessing the OCH1 knock-out and expressing the kringle 3 domain (K3) of human plasminogen, was used as the host strain. BK64 was transformed with the plasmid pPB 103 linearized with the restriction enzyme EcoNI to introduce the K. lactis UDP-N-acetylglucosamine transporter into the host cell, thus creating the strain PBP-1. The mouse Mnsl was introduced into this strain by transformation with the plasmid pFB8 linearized with the restriction enzyme EcoNI, generating strain PBP-2. K3 glycan analysis from proteins isolated from strain PBP-2 demonstrated that the primary glycoform present was Man5GlcNAc2.
  • PBP-2 was subsequently transformed with the human GnTI plasmid pNA15 linearized with the restriction enzyme AatII, generating the strain PBP-3. Analysis of the K3 glycoforms produced in strain PBP-3 demonstrated that the hybrid glycan GlcNAcMan5GlcNAc2 was the predominant structure. To recover the URA3 marker from PBP-3, this strain was grown in YPD prior to selection on minimal media containing 5-Fluoroorotic (5-FOA, BioVectra) and uracil (Boeke et al. (1984) Mol. Gen. Genet. 197:345-346). The recovered Ura-minus strain producing GlcNAcMan5GlcNAc2 glycoforms was designated YSH-1 (FIG. 36). The N-glycan profile from strain YSH-1 is shown in FIG. 25 (top) and displays a predominant peak at 1465 m/z corresponding to the mass of GlcNAcMan5GlcNAc2[d].
  • EXAMPLE 14 Generation of Yeast Strain YSH-37 (P. pastoris Expressing Mannosidase II)
  • YSH-1 (Example 13) was transformed with the D. melanogaster mannosidase IIΔ74/ S. cerevisiae MNN2(s) plasmid (pKD53) linearized with the restriction enzyme ApaI, generating strain YSH-37 (FIG. 36). Analysis of the K3 glycan structures produced in strain YSH-37 (FIG. 25 (bottom)) demonstrated that the predominant glycoform at 1140 m/z corresponds to the mass of GlcNAcMan3GlcNAc2[b] and other glycoforms GlcNAcMan4GlcNAc2[c] at 1303 m/z and GlcNAcMan5GlcNAc2[d] at 1465 m/z.
  • EXAMPLE 15 Generation of Yeast Strain YSH-44
  • Strain YSH-37 (Example 14) was transformed with a plasmid encoding a rat GnTII/MNN2 (s) leader, pTC53, linearized with the restriction enzyme EcoRI. The resulting strain, YSH-44 (FIG. 36), produced a K3 N-glycan having a single glycoform at 1356 m/z, corresponding to the mass of GlcNAc2Man3GlcNAc2[x], by positive mode MALDI-TOF mass spectrometry (FIG. 29).
  • EXAMPLE 16 Construction of Plasmid pJN 348
  • The plasmid pBLURA-SX (from Jim Cregg) was digested with BamHI and BglII to release the AOXexpression cassette. The BamHI fragment containing the GAPDH/CYC1 expression cassette from pJN261 (FIG. 4B) (Example 4) was then ligated into the pBLURA-SX backbone to create pJN338. The plasmid pJN338 was cut with NotI and PacI and the two oligonucleotides 5′-GGCCGCCTGCAGATTTAAATGAATTCGGCGCGCCTTAAT-3′ (SEQ ID NO:96) and 5′-TAAGGCGCGCC GAATTCATTTAAATCTGCAGGGC-3′ (SEQ ID NO:97) that had been annealed in vitro, were ligated into the open sites, to create pJN348.
  • EXAMPLE 17 Construction of an Integration Plasmid pRCD259
  • The PpURA3 containing GAPDH expression vector pJN348 was linearized with XhoI and blunted with T4 DNA polymerase and calf intestinal phosphatase (CIP) treated. The HYG resistance marker was digested from pAG32 with BglII and SacI and blunted, then ligated into pJN348 to create pRCD259 which can be used as a HYG expression vector that integrates at the PpURA3 locus.
  • EXAMPLE 18 Generation of GnTIII Fusion Constructs
  • Fusion constructs between mammalian GnTIII and yeast targeting sequences were made using mouse Mgat3 gene (GenBank accession number L39373, Bhaumik et al., 1995). Three DNA fragments corresponding to N-terminal deletions Δ32, Δ86, and Δ212 of the mouse GnTIII gene were PCR amplified using Pfu Turbo polymerase (Stratagene) with forward MG3-B (5′-TCCTGGCGCGCCTTCCCGAGAGAACTGGCCTCCCTC-3′) (SEQ ID NO:98), MG3-C (5′-CCGAGGCGCGCCACAGAGGAACTGCACCGGGTG-3′) (SEQ ID NO:99), MG3 -D (5′-ACCGAGGCGCGCCATCAACGCCATCAACATCAACCAC-3′) (SEQ ID NO:I00), and reverse MG3-A (5′-AATTAATTAACCCTAGCCCTCCGCTGTATCCAACTTG-3′) (SEQ ID NO:101) primers. The PCR products were then cloned into pJN 348 vector as AscI-Pacl fragments and sequenced. The resulting vectors pVA (GnTIII Δ32), pVB (GnTIII Δ86), and pVC (GnTIII Δ212) were digested with NotI-AscI enzymes and used for the ligation with yeast leader library (leaders 20-67). These targeting peptides are fused to the catalytic domains selected from the mouse GnTIII with 32, 86, 212 amino acid N-terminal deletions. For example, the MNN2 targeting peptide from S. cerevisiae (long, medium and short) and GNT1 from K. lactis (short, and medium) (see Example 11) are shown in Table 11.
    TABLE 11
    A representative combinatorial library of targeting peptide
    sequences/catalytic domains exhibiting UDP-N-
    Acetylglucosaminyltransferase III (GnTIII) activity in P. pastoris YSH-1
    Targeting peptide
    S. cerevisiae S. cerevisiae S. cerevisiae K. lactis
    MNN2 (s) MNN2 (m) MNN2 (I) GNT1 (m)
    Catalytic Domian Mouse GnTIII 50% 30-40% 20-30% 0%
    Δ32 (pVA53) (pVA54) (pVA55) (pVA51)
    Mouse GnTIII 20-30% 30-40% 20-30% 0%
    Δ86 (pVB53) (pVB54) (pVB55) (pVB51)
    Mouse GnTIII  0% 0% 0% 0%
    Δ212 (pVC53) (pVC54) (pVC55) (pVC51)
  • EXAMPLE 19 Engineering of P. pastoris to Produce Bisected GlcNAc2Man5GlcNAc2
  • The P. pastoris strain producing GlcNAcMan5GlcNAc2 (PBP-3) (see Example 8) was counterselected on 5-FOA, thereby selecting for loss of the URA3+ marker and a ura3− phenotype. This strain, designated YSH-1 (FIG. 36), was transformed with the library of N-acetylglucosaminyltransferase III (GnTIII) catalytic domains (vectors pVA, pVB, and pVC) and leaders. Transformants were grown at 30° C. in BMGY to an OD600 of about 10, harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot. The robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3). The N-glycans were released by PNGase digestion (Example 3). The N-glycans were analyzed with a MALDI-TOF MS (Example 3). The GnTIII activities are shown in Table 11. The number of (+)s, as used herein, indicates the relative levels of bisected N-glycan production of % neutral glycans. Targeting peptide sequences were selected from selected from the group consisting of: Saccharomyces GLS1, Saccharomyces MNS1, Saccharomyces SEC12, Pichia SEC, Pichia OCH 1, Saccharomyces MNN9, Saccharomyces VAN 1, Saccharomyces ANP1, Saccharomyces HOC1, Saccharomyces MNN10, Saccharomyces MNN11, Saccharomyces MNT1, Pichia D2, Pichia D9, Pichia J3, Saccharomyces KTR1, Saccharomyces KTR2, Kluyveromyces GnTI, Saccharomyces MNN2, Saccharomyces MNN5, Saccharomyces YUR1, Saccharomyces MNN1, and Saccharomyces MNN6. The pVA53 transformants exhibiting the bisecting GlcNAc (e.g. GlcNAC2MansGlcNAC2) were designated PBP26 (FIG. 36).
  • EXAMPLE 20 Engineering of P. pastoris YSH-44 to Produce Bisected GlcNAc3Man3GlcNAc2
  • For the expression of GnTIII in the strain YSH-44 (FIG. 36), GnTIII constructs from vectors pVA53, pVB53, pVA54, and pVB54 were transferred as NotI-PacI fragments into pRCD259 to generate vectors pPB135, pPB137, pPB136, and pPB138. The vectors contain HYG resistance marker and P. pastoris URA3 gene as targeting sequence for genomic integration. Plasmids are linearized with SalI, transformed into strain YSH-44 by electroporation, selected on medium containing hygromycin and the resulting strains are screened by analysis of the released glycans from purified K3. Transformants were grown at 24° C. in BMGY to an OD600 of about I0, harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well forrnat on a Beckman BioMek 2000 laboratory robot (Example 3). The robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3). The N-glycans were released by PNGase digestion. The N-glycans were analyzed with a MALDI-TOF MS (Example 3). The pPB135 transformants exhibiting the bisecting GlcNAc (e.g. GlcNAc2Man5GlcNAc2) were designated YSH-57 (FIG. 36). Table 11 depicts the activity of the mouse GnTIII.
  • EXAMPLE 21 Engineering of P. pastoris PBP6-5 to Produce Bisected GlcNAc3Man3GlcNAc2
  • The P. pastoris PBP6-5 (Example 11) was transformed with the plasmid pPB135 (Table 11) encoding a mouse GnTIII catalytic domain (Δ32) ligated in frame to a targeting peptide derived from S. cerevisiae MNN2. Transformants were grown at 30° C. in BMGY to an OD600 of about 10, harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot. The robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3). The N-glycans were released by PNGase digestion (Example 3). The N-glycans were analyzed with a MALDI-TOF MS (Example 3). Transformants exhibiting the bisecting GlcNAc (e.g. GlcNAc2Man3GlcNAc2) were designated PBP-38 (FIG. 36). Table 11 depicts the activity of the mouse GnTIII.
  • EXAMPLE 22 In vitro GnTIII Activity Assay using Substrate GlcNAcMan5GlcNAc2 in Engineered P. pastoris Strain YSH-57
  • To test any potential ex vivo GnTIII activity in the P. pastoris strain, YSH-57 cell culture supernatants were tested for GnTIII activity. P. pastoris YSH-57 cells were grown at 24° C. in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for GnTIII assays and the remainder was used for the recovery of secreted soluble K3. K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot. The robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3). The N-glycans were released by PNGase digestion (Example 3). The earlier removed aliquot of the supernatant was further tested for the presence of secreted GnTIII activity. GlcNAcMan5GlcNAc2 purified from K3 expressed in PBP-3 strain was added to: BMMY (A) 1 mM UDP-GlcNAc (Sigma Chemical Co., St. Louis, Mo.)) in BMMY (B); the supernatant of YSH-44 transformed with pVA53 [YSH-57] (C); the supernatant of YSH-57+1 mM UDP-GlcNAc (D). After incubation for 8 hours at room temperature, samples were analyzed by amino silica HPLC to determine the extent of GnTIII activity.
  • EXAMPLE 23 In vitro GnTIII Activity Assay using Substrate GlcNAc2Man3GlcNAc2 in Engineered P. pastoris Strain YSH-57
  • To test any potential ex vivo GnTIII activity in the P. pastoris strain YSH-57 cell culture supernatants were tested for GnTIII activity. P. pastoris YSH-57 cells were grown at 24° C. in BMGY to an OD600 of about 10. Cells were harvested by centrifugation and transferred to BMMY to induce the production of K3 (kringle 3 from human plasminogen) under control of an AOX1 promoter. After 24 hours of induction, cells were removed by centrifugation to yield an essentially clear supernatant. An aliquot of the supernatant was removed for GnTIII assays and the remainder was used for the recovery of secreted soluble K3. K3 was purified from the medium by Ni-affinity chromatography utilizing a 96-well format on a Beckman BioMek 2000 laboratory robot. The robotic purification is an adaptation of the protocol provided by Novagen for their HisBind resin (Example 3). The N-glycans were released by PNGase digestion (Example 3). The earlier removed aliquot of the supernatant was further tested for the presence of secreted GnTIII activity. GlcNAc2Man3GlcNAc2 purified from K3 expressed in YSH-44 strain was added to: BMMY (A) 1 mM UDP-GlcNAc (Sigma Chemical Co., St. Louis, Mo.)) in BMMY (B); the supernatant of YSH-44 transformed with pVA53 [YSH-57] (C). After incubation for 8 hours at room temperature, samples were analyzed by amino silica HPLC to determine the extent of GnTIII activity.

Claims (54)

1. A process of making a human-like glycoprotein in a lower eukaryotic host cell comprising the step of introducing into the cell an N-acetylglucosaminyltransferase III activity.
2. A process of making a human-like glycoprotein in a lower eukaryotic host cell comprising the step of expressing in the cell an N-acetylglucosaminyltransferase III activity.
3. A process of making a human-like glycoprotein in a lower eukaryotic host cell comprising the step of expressing in the cell one or more enzymatic activities that produce N-glycans comprising GlcNAc3Man3GlcNAc2, GlcNAc2Man3GlcNAc2 or GlcNAc2Man5GlcNAc2 bisected structures.
4. The process of claims 1 or 2, wherein the N-acetylglucosaminyltransferase III activity produces a bisected glycan.
5. The process of claims 1 or 2, wherein the glycoprotein comprises a bisected glycan.
6. The process of claims 1 or 2, wherein the activity is substantially intracellular.
7. The process of claims 1, 2, or 3, further comprising the step of isolating the glycoprotein from the host cell.
8. The process of claims 1, 2, or 3, wherein the host cell is selected from the group consisting of Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium lucknowense, Fusarium sp., Fusarium gramineum, Fusarium venenatum, and Neurospora crassa.
9. The process of claim 8, wherein the host cell is selected from the group consisting of Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, and Pichia sp.
10. The process of claim 9, wherein the host cell is Pichia pastoris.
11. The process of claims 1, 2, or 3, wherein the glycoprotein is a therapeutic protein.
12. The process of claim 11, wherein the therapeutic protein is selected from the group consisting of erythropoietin, cytokines, coagulation factors, soluble IgE receptor a-chain, IgG, IgG fragments, IgM, interleukins, urokinase, chymase, urea trypsin inhibitor, IGF-binding protein, epidermal growth factor, growth hormone-releasing factor, annexin V fusion protein, angiostatin, vascular endothelial growth factor-2, myeloid progenitor inhibitory factor-1, osteoprotegerin, α-1-antitrypsin, α-feto protein, and DNase II.
13. A lower eukaryotic host cell comprising an N-acetylglucosaminyltransferase III activity.
14. A lower eukaryotic host cell comprising an N-acetylglucosaminyltransferase II activity and an N-acetylglucosaminyltransferase III activity.
15. The host cell of claim 13 or 14, wherein the activity is substantially intracellular.
16. The host cell of claim 13 or 14, wherein the cell produces N-glycans comprising GlcNAcMan3GlcNAc2 structures that are capable of reacting with GnTIII activity.
17. The host cell of claim 13 or 14, wherein the N-acetylglucosaminyltransferase III activity produces a bisected glycan.
18. A lower eukaryotic host cell comprising an N-glycan that comprises a bisected glycan.
19. The host cell of claim 18, wherein the N-glycan contains greater than 10 mole % of the bisected glycan.
20. The host cell of claim 18, wherein the N-glycan contains greater than 80 mole % of the bisected glycan.
21. A lower eukaryotic host cell comprising an N-glycan that comprises GlcNAc3Man3GlcNAc2, GlcNAc2Man3GlcNAc2 or GlcNAc2Man5GlcNAc2 bisected structures.
22. The host cell of claims 13, 14, 18, or 21, wherein the host cell is selected from the group consisting of Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium lucknowense, Fusarium sp., Fusarium gramineum, Fusarium venenatum, and Neurospora crassa.
23. The host cell of claim 22, wherein the host cell is selected from the group consisting of Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, and Pichia sp..
24. The host cell of claim 23, wherein the host cell is Pichia pastoris.
25. A lower eukaryotic host cell comprising a Man5GlcNAc2 core structure or a Man3GlcNAc2 core structure modified by a bisecting GlcNAc.
26. The host cell of claim 25, wherein the cell produces greater than 10 mole % of the modified structure.
27. The host cell of claim 25, wherein the cell produces greater than 50 mole % of the modified structure.
28. A lower eukaryotic host cell comprising an N-acetylglucosaminyltransferase I activity and an N-acetylglucosaminyltransferase III activity.
29. The host cell of claim 28, wherein the activities are substantially intracellular.
30. The host cell of claim 28, wherein the cell produces N-glycans comprising GlcNAcMan3GlcNAc2 that are capable of reacting with GnTIII activity.
31. The host cell of claim 28, wherein the N-acetylglucosaminyltransferase III activity produces a bisected glycan.
32. A lower eukaryotic host cell comprising a GnTIII activity and a mannosidase II activity.
33. The host cell of claim 32, further comprising an N-acetylglucosaminyltransferase I activity.
34. The host cell of claim 32, further comprising an N-acetylglucosaminyltransferase II activity.
35. The host cell of claim 32, further comprising an N-acetylglucosaminyltransferase I activity and an N-acetylglucosaminyltransferase II activity.
36. The host cell of claims 13, 14, 28, or 32 that is deficient in an OCH1 mannosyltransferase activity.
37. The host cell of claims 13, 14, 28, or 32 that is deficient in a Do1-P-Man:Man5GlcNAc2-PP-Do1 mannosyltransferase activity.
38. The host cell of claims 13, 14, 28, or 32, further comprising an α-1,2-mannosidase I activity.
39. The host cell of claims 13, 14, 28, or 32, further comprising a UDP-GlcNAc transporter.
40. The glycoprotein made by the process of claim 1.
41. The glycoprotein made by the process of claim 2.
42. The glycoprotein made by the process of claim 3.
43. The glycoprotein made by the process of claim 8.
44. The glycoprotein made by the process of claim 9.
45. The glycoprotein made by the process of claim 10.
46. The glycoprotein made by the process of claim 11.
47. The glycoprotein made by the process of claim 12.
48. A glycoprotein comprising a bisecting GlcNAc on a Man5GlcNAc2 or a Man3GlcNAc2 core structure produced in a lower eukaryotic host cell.
49. A glycoprotein comprising a bisecting GlcNAc attached to a Man5GlcNAc2, Man4GlcNAc2, Man3GlcNAc2, GlcNAcMan3GlcNAc2, GlcNAcMan5GlcNAc2, or a GlcNAc2Man3GlcNAc2 core structure produced in a lower eukaryotic host cell.
50. The glycoprotein of claim 49, wherein greater than 10 mole % of the core structures are modified by the bisecting GlcNAc.
51. The glycoprotein of claim 49, wherein greater than 80 mole % of the core structures are modified by the bisecting GlcNAc.
52. A pharmaceutical composition comprising the glycoprotein of any one of claims 40-51.
53. A vector selected from the group consisting of pVA, pVB, and pVC.
54. The vector of claim 53, wherein the vector is selected from the group consisting of pVA53, pVA54, pVA55, pVB53, pVB54, and pVB55.
US10/680,963 2000-06-28 2003-10-07 N-acetylglucosaminyltransferase III expression in lower eukaryotes Expired - Fee Related US7598055B2 (en)

Priority Applications (33)

Application Number Priority Date Filing Date Title
US10/680,963 US7598055B2 (en) 2000-06-28 2003-10-07 N-acetylglucosaminyltransferase III expression in lower eukaryotes
PT04713412T PT1599595E (en) 2003-02-20 2004-02-20 N -acetylglucosaminyltransferase iii expression in lower eukaryotes
ES09004760T ES2384929T3 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins that have multiantennial structures
DE602004024289T DE602004024289D1 (en) 2003-02-20 2004-02-20 N-ACETYLGLUCOSAMINE TRANSFERASE III EXPRESSION IN LOW EUKARYOTES
ES04713388T ES2336790T5 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins with multiple antennial structures
EP04713412.7A EP1599595B2 (en) 2003-02-20 2004-02-20 N -acetylglucosaminyltransferase iii expression in lower eukaryotes
SI200431362T SI1597381T2 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
AT04713412T ATE449862T1 (en) 2003-02-20 2004-02-20 N-ACETYLGLUCOSAMINYLTRANSFERASE III EXPRESSION IN LOW EUKARYOTES
EP04713388.9A EP1597381B2 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
CA2516550A CA2516550C (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
AT04713388T ATE452201T1 (en) 2003-02-20 2004-02-20 PRODUCTION OF MODIFIED GLYCOPROTEINS WITH MULTIPLY BRANCHED STRUCTURES
AU2004213868A AU2004213868B2 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
US10/546,101 US8697394B2 (en) 2000-06-28 2004-02-20 Production of modified glycoproteins having multiple antennary structures
CA2753408A CA2753408A1 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
DE602004024649T DE602004024649D1 (en) 2003-02-20 2004-02-20 PRODUCTION OF MODIFIED GLYCOPROTEINS WITH MULTIPLE BRANCHED STRUCTURES
CA2516520A CA2516520C (en) 2003-02-20 2004-02-20 N-acetylglucosaminyltransferase iii expression in lower eukaryotes
AT09004760T ATE543912T1 (en) 2003-02-20 2004-02-20 PRODUCTION OF MODIFIED GLYCOPROTEINS WITH MULTIANTENNARY STRUCTURES
JP2006503776A JP4758335B2 (en) 2003-02-20 2004-02-20 Generation of modified glycoproteins with multiple antenna structures
SI200431310T SI1599595T2 (en) 2003-02-20 2004-02-20 N -acetylglucosaminyltransferase iii expression in lower eukaryotes
PCT/US2004/005191 WO2004074461A2 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
PCT/US2004/005128 WO2004074458A2 (en) 2003-02-20 2004-02-20 N-acetylglucosaminyltransferase iii expression in lower eukaryotes
ES04713412T ES2335345T5 (en) 2003-02-20 2004-02-20 Expression of N-acetylglucosaminyltransferase III in lower eukaryotes
PT04713388T PT1597381E (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
JP2006503757A JP4611280B2 (en) 2003-02-20 2004-02-20 N-acetylglucosaminyltransferase III expression in lower eukaryotes
AU2004213859A AU2004213859B2 (en) 2003-02-20 2004-02-20 N-acetylglucosaminyltransferase III expression in lower eukaryotes
EP09004760A EP2080809B1 (en) 2003-02-20 2004-02-20 Production of modified glycoproteins having multiple antennary structures
US12/313,636 US20090226959A1 (en) 2000-06-28 2008-11-21 Production of modified glycoproteins having multiple antennary structures
US12/540,849 US8445227B2 (en) 2000-06-28 2009-08-13 N-acetylglucosaminyltransferase III expression in lower eukaryotes
US12/540,915 US20100016555A1 (en) 2000-06-28 2009-08-13 N-Acetylglucosaminyltransferase III Expression in Lower Eukaryotes
AU2010201036A AU2010201036B2 (en) 2003-02-20 2010-03-17 Production of Modified Glycoproteins Having Multiple Antennary Structures
JP2010104525A JP5255017B2 (en) 2003-02-20 2010-04-28 Generation of modified glycoproteins with multiple antenna structures
JP2010181319A JP2011019521A (en) 2003-02-20 2010-08-13 N-acetylglucosaminyltransferase iii expression in lower eukaryote
US14/340,621 US20150079633A1 (en) 2000-06-28 2014-07-25 Production of modified glycoproteins having multiple antennary structures

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US21435800P 2000-06-28 2000-06-28
US21563800P 2000-06-30 2000-06-30
US27999701P 2001-03-30 2001-03-30
US09/892,591 US7029872B2 (en) 2000-06-28 2001-06-27 Methods for producing modified glycoproteins
US34416901P 2001-12-27 2001-12-27
PCT/US2002/041510 WO2003056914A1 (en) 2001-12-27 2002-12-24 Methods to engineer mammalian-type carbohydrate structures
US10/371,877 US7449308B2 (en) 2000-06-28 2003-02-20 Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US10/680,963 US7598055B2 (en) 2000-06-28 2003-10-07 N-acetylglucosaminyltransferase III expression in lower eukaryotes

Related Parent Applications (4)

Application Number Title Priority Date Filing Date
PCT/US2002/041510 Continuation-In-Part WO2003056914A1 (en) 2000-06-28 2002-12-24 Methods to engineer mammalian-type carbohydrate structures
US10/500,240 Continuation-In-Part US8932825B2 (en) 2001-12-27 2002-12-24 Method to engineer mammalian-type carbohydrate structures
US10/371,877 Continuation-In-Part US7449308B2 (en) 2000-06-28 2003-02-20 Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US50024005A Continuation-In-Part 2000-06-28 2005-03-23

Related Child Applications (6)

Application Number Title Priority Date Filing Date
US10/371,877 Continuation-In-Part US7449308B2 (en) 2000-06-28 2003-02-20 Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US10/546,101 Continuation-In-Part US8697394B2 (en) 2000-06-28 2004-02-20 Production of modified glycoproteins having multiple antennary structures
PCT/US2004/005191 Continuation-In-Part WO2004074461A2 (en) 2000-06-28 2004-02-20 Production of modified glycoproteins having multiple antennary structures
US11/546,101 Continuation-In-Part US7673796B2 (en) 2005-11-28 2006-10-11 System and method for providing remote site intervention support for self-checkout stations
US12/540,849 Division US8445227B2 (en) 2000-06-28 2009-08-13 N-acetylglucosaminyltransferase III expression in lower eukaryotes
US12/540,915 Continuation US20100016555A1 (en) 2000-06-28 2009-08-13 N-Acetylglucosaminyltransferase III Expression in Lower Eukaryotes

Publications (2)

Publication Number Publication Date
US20050208617A1 true US20050208617A1 (en) 2005-09-22
US7598055B2 US7598055B2 (en) 2009-10-06

Family

ID=32911943

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/680,963 Expired - Fee Related US7598055B2 (en) 2000-06-28 2003-10-07 N-acetylglucosaminyltransferase III expression in lower eukaryotes
US12/540,849 Expired - Fee Related US8445227B2 (en) 2000-06-28 2009-08-13 N-acetylglucosaminyltransferase III expression in lower eukaryotes
US12/540,915 Abandoned US20100016555A1 (en) 2000-06-28 2009-08-13 N-Acetylglucosaminyltransferase III Expression in Lower Eukaryotes

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/540,849 Expired - Fee Related US8445227B2 (en) 2000-06-28 2009-08-13 N-acetylglucosaminyltransferase III expression in lower eukaryotes
US12/540,915 Abandoned US20100016555A1 (en) 2000-06-28 2009-08-13 N-Acetylglucosaminyltransferase III Expression in Lower Eukaryotes

Country Status (11)

Country Link
US (3) US7598055B2 (en)
EP (3) EP2080809B1 (en)
JP (4) JP4611280B2 (en)
AT (3) ATE543912T1 (en)
AU (3) AU2004213868B2 (en)
CA (3) CA2516550C (en)
DE (2) DE602004024649D1 (en)
ES (3) ES2384929T3 (en)
PT (2) PT1599595E (en)
SI (2) SI1597381T2 (en)
WO (2) WO2004074461A2 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040018590A1 (en) * 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US20040230042A1 (en) * 2000-06-28 2004-11-18 Stephen Hamilton Expression of class 2 mannosidase and class III mannosidase in lower eukaryotic cells
US20050170452A1 (en) * 2001-12-27 2005-08-04 Stefan Wildt Method to engineer mammanlian-type carbohydrate structures
US20060024304A1 (en) * 2000-06-28 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Man5GlcNAc2 glycoform
US20060024292A1 (en) * 2001-12-27 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Gal2GlcNAc2Man3GlcNAc2 glycoform
US20060029604A1 (en) * 2000-06-28 2006-02-09 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAc2Man3GlcNAc2 glycoform
US20060034829A1 (en) * 2001-12-27 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a MAN3GLCNAC2 glycoform
US20060034828A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAcMAN5GLCNAC2 glycoform
US20060068035A1 (en) * 2002-03-07 2006-03-30 Pero Ronald W Method of preparation and composition of a water soluble extract of the bioactive component of the plant species Uncaria for enhancing immune, anti-inflammatory, anti-tumor and DNA repair processes of warm blooded animals
US20060078963A1 (en) * 2000-06-28 2006-04-13 Glycofi, Inc. Methods for producing modified glycoproteins
US20060286637A1 (en) * 2000-06-28 2006-12-21 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US20080085540A1 (en) * 2004-03-17 2008-04-10 Hamilton Stephen R Method of engineering a cytidine monophosphate-sialic acid synthetic pathway in fungi and yeast
US20090136525A1 (en) * 2005-09-02 2009-05-28 Tillman Gerngross Immunoglobulins Comprising Predominantly a Glcnacman3Glcnac2 Glycoform
US20090170159A1 (en) * 2005-11-15 2009-07-02 Glycofi, Inc Production of glycoproteins with reduced o-glycosylation
US20090226464A1 (en) * 2005-09-09 2009-09-10 Tillman Gerngross Immunoglobulins comprising predominantly a glcnacman3glcnac2 glycoform
US7598055B2 (en) 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
US20100028951A1 (en) * 2007-03-07 2010-02-04 Stephen Hamilton Production of glycoproteins with modified fucosylation
US20100062485A1 (en) * 2005-10-27 2010-03-11 Korea Research Institute Of Bioscience And Biotechnology Novel Hansenula Polymorpha Gene Coding for Dolichyl-Phosphate- Mannose Dependent Alpha-1,3 Mannosyltransferase and Process for the Production of Recombinant Glycoproteins With Hansenula Polymorpha Mutant Strain Deficient in
WO2011106389A1 (en) 2010-02-24 2011-09-01 Merck Sharp & Dohme Corp. Method for increasing n-glycosylation site occupancy on therapeutic glycoproteins produced in pichia pastoris
US20120283420A1 (en) * 2009-04-22 2012-11-08 Bieke Nagels Production of Multi-Antennary N-Glycan Structures in Plants
EP2546347A2 (en) 2009-02-25 2013-01-16 Merck Sharp & Dohme Corp. Glycoprotein composition from engineered galactose assimilation pathway in Pichia pastoris
WO2014064203A1 (en) 2012-10-26 2014-05-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Lyve-1 antagonists for preventing or treating a pathological condition associated with lymphangiogenesis
US8741633B2 (en) 2006-05-19 2014-06-03 Glycofi, Inc. Recombinant vectors
WO2014090948A1 (en) 2012-12-13 2014-06-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Serpin spn4a and biologically active derivatives thereof for use in the treatment of cancer
WO2014111467A1 (en) 2013-01-16 2014-07-24 INSERM (Institut National de la Santé et de la Recherche Médicale) A soluble fibroblast growth factor receptor 3 (fgr3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
WO2015044379A1 (en) 2013-09-27 2015-04-02 INSERM (Institut National de la Santé et de la Recherche Médicale) A dyrk1a polypeptide for use in preventing or treating metabolic disorders
US9328170B2 (en) 2011-05-25 2016-05-03 Merck Sharp & Dohme Corp. Method for preparing Fc containing polypeptides having improved properties
US9695454B2 (en) 2012-05-23 2017-07-04 Glykos Finland Oy Production of fucosylated glycoproteins
US10513724B2 (en) 2014-07-21 2019-12-24 Glykos Finland Oy Production of glycoproteins with mammalian-like N-glycans in filamentous fungi
CN112041456A (en) * 2018-05-01 2020-12-04 科纳根公司 Recombinant organisms and methods for producing sugar molecules with low sulfation
US10858686B2 (en) 2010-05-27 2020-12-08 Merck Sharp & Dohme Corp. Method for preparing antibodies having improved properties
EP4309666A1 (en) 2022-07-21 2024-01-24 Technische Universität Dresden M-csf for use in the prophylaxis and/or treatment of viral infections in states of immunosuppression
WO2024017998A1 (en) 2022-07-21 2024-01-25 Technische Universität Dresden M-csf for use in the prophylaxis and/or treatment of viral infections in states of immunosuppression
US11959118B2 (en) 2020-10-20 2024-04-16 Merck Sharp & Dohme Llc Fc-containing polypeptides having improved properties and comprising mutations at positions 243 and 264 of the Fc-region

Families Citing this family (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US8697394B2 (en) * 2000-06-28 2014-04-15 Glycofi, Inc. Production of modified glycoproteins having multiple antennary structures
HUP0600342A3 (en) * 2001-10-25 2011-03-28 Genentech Inc Glycoprotein compositions
CA2483436A1 (en) 2002-04-23 2003-11-06 University Of Georgia Research Foundation, Inc. N-acetylglucosaminyltransferase vb coding sequence, recombinant cells and methods
WO2004046358A1 (en) 2002-11-21 2004-06-03 Chugai Seiyaku Kabushiki Kaisha Novel cancer-associated gene
US7332299B2 (en) 2003-02-20 2008-02-19 Glycofi, Inc. Endomannosidases in the modification of glycoproteins in eukaryotes
US7507573B2 (en) * 2003-11-14 2009-03-24 Vib, Vzw Modification of protein glycosylation in methylotrophic yeast
GB0513881D0 (en) 2005-07-06 2005-08-10 Btg Int Ltd Core 2 GLCNAC-T Inhibitors III
GB0329667D0 (en) 2003-12-22 2004-01-28 King S College London Core 2 GlcNAc-T inhibitor
US20080182801A1 (en) 2003-12-22 2008-07-31 Btg International Limited Core 2 glcnac-t inhibitors
GB0507123D0 (en) * 2005-04-08 2005-05-11 Isis Innovation Method
GB0513888D0 (en) 2005-07-06 2005-08-10 Btg Int Ltd Core 2 GLCNAC-T Inhibitors II
WO2007035930A2 (en) * 2005-09-22 2007-03-29 Prosci Incorporated Glycosylated polypeptides produced in yeast mutants and methods of use thereof
EP1974040B1 (en) * 2006-01-17 2012-10-03 Biolex Therapeutics, Inc. Compositions and methods for humanization and optimization of N-glycans in plants
MX2008014744A (en) * 2006-05-19 2009-02-10 Glycofi Inc Erythropoietin compositions.
ATE516346T1 (en) 2006-05-24 2011-07-15 Univ Provence Aix Marseille 1 PREPARATION AND USES OF CHIMERIC GLYCOSYL TRANSFERASES WITH GENE SEQUENCES ENCODING OPTIMIZED GLYCOSYLATION ACTIVITY
RU2518289C2 (en) 2006-09-13 2014-06-10 Эббви Инк, Method for preparing fed-batch antibody or its fragment (versions)
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
US20080127996A1 (en) * 2006-12-04 2008-06-05 Weinhold Dennis G Method and apparatus to remediate an acid and/or liquid spill
KR101811057B1 (en) * 2007-04-03 2017-12-20 옥시레인 유케이 리미티드 Glycosylation of molecules
EP1995312A1 (en) * 2007-05-23 2008-11-26 Plant Research International B.V. Alg3 mutant
SG195558A1 (en) * 2008-10-14 2013-12-30 Genentech Inc Immunoglobulin variants and uses thereof
CN104974251A (en) 2008-10-20 2015-10-14 Abbvie公司 Viral inactivation during purification of antibodies
WO2010141039A1 (en) 2008-10-20 2010-12-09 Abbott Laboratories Isolation and purification of antibodies using protein a affinity chromatography
US20110135630A1 (en) * 2009-07-13 2011-06-09 Tolbert Thomas J Chemical modification of antibody fragments
CN102712914A (en) 2009-09-29 2012-10-03 根特大学 Hydrolysis of mannose-1-phospho-6-mannose linkage to phospho-6-mannose
WO2011053541A1 (en) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Methods for the production of recombinant proteins with improved secretion efficiencies
CN102834509A (en) * 2009-11-19 2012-12-19 奥克西雷恩英国有限公司 Yeast strains producing mammalian-like complex N-glycans
BR112013007263A2 (en) 2010-09-29 2016-06-14 Oxyrane Uk Ltd demanosylation of phosphorylated n-glycans
US9347050B2 (en) 2010-09-29 2016-05-24 Oxyrane Uk Limited Mannosidases capable of uncapping mannose-1-phospho-6-mannose linkages and demannosylating phosphorylated N-glycans and methods of facilitating mammalian cellular uptake of glycoproteins
DK2643456T3 (en) * 2010-11-24 2016-08-22 Glykos Finland Oy FUSION enzymes with N-ACETYLGLUKOSAMINYLTRANSFERASE ACTIVITY
WO2012115903A2 (en) 2011-02-25 2012-08-30 Merck Sharp & Dohme Corp. Yeast strain for the production of proteins with modified o-glycosylation
MY163539A (en) 2011-03-29 2017-09-15 Roche Glycart Ag Antibody fc variants
WO2012149197A2 (en) 2011-04-27 2012-11-01 Abbott Laboratories Methods for controlling the galactosylation profile of recombinantly-expressed proteins
JP2014527518A (en) 2011-07-20 2014-10-16 ゼプテオン インコーポレイテッド Polypeptide separation method
IN2014CN02964A (en) * 2011-10-28 2015-07-03 Merck Sharp & Dohme
BR112014022624A2 (en) 2012-03-15 2017-07-11 Oxyrane Uk Ltd Methods and Materials for Treatment of Pompe Disease
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
CA2874554C (en) 2012-07-13 2019-12-03 Roche Glycart Ag Bispecific anti-vegf/anti-ang-2 antibodies and their use in the treatment of ocular vascular diseases
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
CA2883272A1 (en) 2012-09-02 2014-03-06 Abbvie Inc. Methods to control protein heterogeneity
KR102134936B1 (en) * 2012-10-22 2020-07-16 머크 샤프 앤드 돔 코포레이션 Crz1 mutant fungal cells
SG11201507230PA (en) 2013-03-12 2015-10-29 Abbvie Inc Human antibodies that bind human tnf-alpha and methods of preparing the same
CA2916421A1 (en) 2013-03-14 2014-09-25 Bayer Healthcare Llc Monoclonal antibodies against antithrombin beta complexed with heparin
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
WO2015051293A2 (en) 2013-10-04 2015-04-09 Abbvie, Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
WO2016077457A1 (en) 2014-11-11 2016-05-19 Clara Foods Co. Methods and compositions for egg white protein production
CA2977271A1 (en) 2015-03-02 2016-09-09 Synthetic Genomics, Inc. Regulatory elements from labyrinthulomycetes microorganisms
WO2017191208A1 (en) * 2016-05-03 2017-11-09 Vib Vzw Means and methods for generating complex glycans derived from fungal engineered hosts
US10633454B2 (en) 2016-11-01 2020-04-28 Conagen Inc. Expression of modified glycoproteins and glycopeptides
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
KR20220034848A (en) 2019-07-11 2022-03-18 클라라 푸드즈 컴퍼니 Protein composition and edible products thereof
US10927360B1 (en) 2019-08-07 2021-02-23 Clara Foods Co. Compositions comprising digestive enzymes

Citations (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4414329A (en) * 1980-01-15 1983-11-08 Phillips Petroleum Company Biochemical conversions by yeast fermentation at high cell densities
US4617274A (en) * 1981-10-29 1986-10-14 Phillips Petroleum Company Biochemical conversions by yeast fermentation at high cell densities
US4683293A (en) * 1986-10-20 1987-07-28 Phillips Petroleum Company Purification of pichia produced lipophilic proteins
US4775622A (en) * 1982-03-08 1988-10-04 Genentech, Inc. Expression, processing and secretion of heterologous protein by yeast
US4808537A (en) * 1984-10-30 1989-02-28 Phillips Petroleum Company Methanol inducible genes obtained from pichia and methods of use
US4812405A (en) * 1986-02-18 1989-03-14 Phillips Petroleum Company Double auxotrophic mutants of Pichia pastoris and methods for preparation
US4818700A (en) * 1985-10-25 1989-04-04 Phillips Petroleum Company Pichia pastoris argininosuccinate lyase gene and uses thereof
US4837148A (en) * 1984-10-30 1989-06-06 Phillips Petroleum Company Autonomous replication sequences for yeast strains of the genus pichia
US4855231A (en) * 1984-10-30 1989-08-08 Phillips Petroleum Company Regulatory region for heterologous gene expression in yeast
US4857467A (en) * 1986-07-23 1989-08-15 Phillips Petroleum Company Carbon and energy source markers for transformation of strains of the genes Pichia
US4879231A (en) * 1984-10-30 1989-11-07 Phillips Petroleum Company Transformation of yeasts of the genus pichia
US4882279A (en) * 1985-10-25 1989-11-21 Phillips Petroleum Company Site selective genomic modification of yeast of the genus pichia
US4885242A (en) * 1984-10-30 1989-12-05 Phillips Petroleum Company Genes from pichia histidine pathway and uses thereof
US4925796A (en) * 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US4929555A (en) * 1987-10-19 1990-05-29 Phillips Petroleum Company Pichia transformation
US4935349A (en) * 1986-01-17 1990-06-19 Zymogenetics, Inc. Expression of higher eucaryotic genes in aspergillus
US5002876A (en) * 1986-09-22 1991-03-26 Phillips Petroleum Company Yeast production of human tumor necrosis factor
US5004688A (en) * 1988-04-15 1991-04-02 Phillips Petroleum Company Purification of hepatitis proteins
US5032519A (en) * 1989-10-24 1991-07-16 The Regents Of The Univ. Of California Method for producing secretable glycosyltransferases and other Golgi processing enzymes
US5032516A (en) * 1985-10-25 1991-07-16 Phillips Petroleum Company Pichia pastoris alcohol oxidase II regulatory region
US5047335A (en) * 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5122465A (en) * 1989-06-12 1992-06-16 Phillips Petroleum Company Strains of pichia pastoris created by interlocus recombination
US5135854A (en) * 1987-10-29 1992-08-04 Zymogenetics, Inc. Methods of regulating protein glycosylation
US5166329A (en) * 1985-10-25 1992-11-24 Phillips Petroleum Company DNA encoding the alcohol oxidase 2 gene of yeast of the genus Pichia
US5324663A (en) * 1990-02-14 1994-06-28 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US5595900A (en) * 1990-02-14 1997-01-21 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US5602003A (en) * 1992-06-29 1997-02-11 University Of Georgia Research Foundation N-acetylglucosaminyltransferase V gene
US5707828A (en) * 1991-04-25 1998-01-13 Research Corporation Technologies, Inc. Expression of human serum albumin in Pichia pastoris
US5766910A (en) * 1993-09-09 1998-06-16 La Jolla Cancer Research Foundation Expression of the developmental I antigen by a cloned human cDNA encoding a member of a beta-1, 6-N-acetylglucosaminyltransfrase gene family
US5834251A (en) * 1994-12-30 1998-11-10 Alko Group Ltd. Methods of modifying carbohydrate moieties
US5849904A (en) * 1994-12-22 1998-12-15 Boehringer Mannheim Gmbh Isolated nucleic acid molecules which hybridize to polysialyl transferases
US5854018A (en) * 1981-02-25 1998-12-29 Washington Research Foundation Expression of polypeptides in yeast
US5861293A (en) * 1994-06-13 1999-01-19 Banyu Pharmaceutical Co., Ltd. Gene encoding glycosyltransferase and its uses
US5910570A (en) * 1995-11-13 1999-06-08 Pharmacia & Upjohn Company Cloned DNA encoding a UDP-GalNAc: polypeptide N-acetylgalactosaminy-ltransferase
US5945314A (en) * 1997-03-31 1999-08-31 Abbott Laboratories Process for synthesizing oligosaccharides
US5945322A (en) * 1994-09-26 1999-08-31 The Rockefeller University Glycosyltransferases for biosynthesis of oligosaccharides, and genes encoding them
US5955422A (en) * 1983-12-13 1999-09-21 Kirin-Amgen, Inc. Production of erthropoietin
US5962294A (en) * 1992-03-09 1999-10-05 The Regents Of The University Of California Compositions and methods for the identification and synthesis of sialyltransferases
US6017743A (en) * 1995-04-03 2000-01-25 The Institute Of Physical And Chemical Research Siaα 2,3, Galβ 1, 4Glc NAcα 2,8-sialyltransferase
US6096512A (en) * 1993-05-14 2000-08-01 Pharmacia & Upjohn Company Cloned DNA encoding a UDP-GalNAc: Polypeptide, N-acetylgalactosaminyltransferase
US6204431B1 (en) * 1994-03-09 2001-03-20 Abbott Laboratories Transgenic non-human mammals expressing heterologous glycosyltransferase DNA sequences produce oligosaccharides and glycoproteins in their milk
US6300113B1 (en) * 1995-11-21 2001-10-09 New England Biolabs Inc. Isolation and composition of novel glycosidases
US6410246B1 (en) * 2000-06-23 2002-06-25 Genetastix Corporation Highly diverse library of yeast expression vectors
US6602684B1 (en) * 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US20030175884A1 (en) * 2001-08-03 2003-09-18 Pablo Umana Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20040018590A1 (en) * 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US20040171826A1 (en) * 2003-02-20 2004-09-02 Stephen Hamilton Endomannosidases in the modification of glycoproteins in eukaryotes
US20040230042A1 (en) * 2000-06-28 2004-11-18 Stephen Hamilton Expression of class 2 mannosidase and class III mannosidase in lower eukaryotic cells
US20050170452A1 (en) * 2001-12-27 2005-08-04 Stefan Wildt Method to engineer mammanlian-type carbohydrate structures
US20050260729A1 (en) * 2004-03-17 2005-11-24 Hamilton Stephen R Method of engineering a cytidine monophosphate-sialic acid synthetic pathway in fungi and yeast
US20060024292A1 (en) * 2001-12-27 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Gal2GlcNAc2Man3GlcNAc2 glycoform
US20060024304A1 (en) * 2000-06-28 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Man5GlcNAc2 glycoform
US20060029604A1 (en) * 2000-06-28 2006-02-09 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAc2Man3GlcNAc2 glycoform
US20060034828A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAcMAN5GLCNAC2 glycoform
US20060034829A1 (en) * 2001-12-27 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a MAN3GLCNAC2 glycoform
US20060034830A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GalGlcNAcMan5GLcNAc2 glycoform
US20060040353A1 (en) * 2000-06-28 2006-02-23 Davidson Robert C Production of galactosylated glycoproteins in lower eukaryotes
US20060078963A1 (en) * 2000-06-28 2006-04-13 Glycofi, Inc. Methods for producing modified glycoproteins
US7064191B2 (en) * 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
US20060160179A1 (en) * 2003-12-24 2006-07-20 Piotr Bobrowicz Methods for eliminating mannosylphosphorylation of glycans in the production of glycoproteins
US20060211085A1 (en) * 2004-04-29 2006-09-21 Piotr Bobrowicz Methods for reducing or eliminating alpha-mannosidase resistant glycans for the production of glycoproteins
US20060286637A1 (en) * 2000-06-28 2006-12-21 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US7214775B2 (en) * 1999-04-09 2007-05-08 Kyowa Hakko Kogyo Co., Ltd. Method of modulating the activity of functional immune molecules

Family Cites Families (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5272066A (en) 1986-03-07 1993-12-21 Massachusetts Institute Of Technology Synthetic method for enhancing glycoprotein stability
US5683899A (en) * 1994-02-03 1997-11-04 University Of Hawaii Methods and compositions for combinatorial-based discovery of new multimeric molecules
US6069235A (en) 1994-02-23 2000-05-30 Monsanto Company Method for carbohydrate engineering of glycoproteins
WO1995025167A1 (en) * 1994-03-17 1995-09-21 Merck Patent Gmbh Anti-egfr single-chain fvs and anti-egfr antibodies
JPH08336387A (en) 1995-06-12 1996-12-24 Green Cross Corp:The Sugar chain-extended protein derived from pichia yeast and dna of the protein
CA2259501A1 (en) * 1996-07-03 1998-01-08 Millennium Pharmaceuticals, Inc. Detection of cancer by assaying for cancer-specific antigens having linked oligosaccharides which are at least triantennary
ATE374816T1 (en) 1996-08-02 2007-10-15 Austin Research Inst IMPROVED NUCLEIC ACIDS ENCODING A CHIMERIC GLYCOSYL TRANSFERASE
US6338955B2 (en) 1996-12-12 2002-01-15 Kirin Beer Kabushiki Kaisha β1-4 N-acetylglucosaminyltransferase and gene encoding
US20040191256A1 (en) * 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US7029870B1 (en) 1997-07-03 2006-04-18 Human Genome Sciences, Inc. Gabaa receptor epsilon subunits
JPH11103158A (en) 1997-09-26 1999-04-13 Olympus Optical Co Ltd Flip-chip mounting to printed wiring board and mounting structure
US7244601B2 (en) 1997-12-15 2007-07-17 National Research Council Of Canada Fusion proteins for use in enzymatic synthesis of oligosaccharides
JPH11221079A (en) 1998-02-04 1999-08-17 Kyowa Hakko Kogyo Co Ltd Transglycosidase and dna encoding the same enzyme
WO1999040208A1 (en) 1998-02-05 1999-08-12 The General Hospital Corporation In vivo construction of dna libraries
EP1211310B1 (en) 1999-08-19 2006-12-13 Kirin Beer Kabushiki Kaisha Novel yeast variants and process for producing glycoprotein containing mammalian type sugar chain
AU7849200A (en) 1999-10-01 2001-05-10 University Of Victoria Innovation And Development Corporation Mannosidases and methods for using same
WO2001029242A2 (en) * 1999-10-21 2001-04-26 Monsanto Company Post-translational modification of recombinant proteins produced in plants
CA2388777A1 (en) 1999-11-19 2001-05-25 Human Genome Sciences, Inc. 18 human secreted proteins
WO2001060860A2 (en) 2000-02-17 2001-08-23 Millennium Predictive Medicine, Inc. Genes differentially expressed in human prostate cancer and their use
US8697394B2 (en) 2000-06-28 2014-04-15 Glycofi, Inc. Production of modified glycoproteins having multiple antennary structures
US20060257399A1 (en) 2000-06-28 2006-11-16 Glycofi, Inc. Immunoglobulins comprising predominantly a Man5GIcNAc2 glycoform
US7598055B2 (en) 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
AU7765801A (en) 2000-06-30 2002-01-08 Flanders Interuniversity Inst Protein glycosylation modification in pichia pastoris
US6946292B2 (en) * 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
JP2002209587A (en) 2001-01-18 2002-07-30 Fujirebio Inc Acetylglucosamine transferase vi protein and nucleic acid encoding the same
JP4655388B2 (en) * 2001-03-05 2011-03-23 富士レビオ株式会社 Protein production method
JP2005500018A (en) * 2001-04-02 2005-01-06 アイデック ファーマスーティカルズ コーポレイション Recombinant antibody coexpressed with GnTIII
CA2448148A1 (en) 2001-05-25 2002-12-05 Incyte Genomics, Inc. Carbohydrate-associated proteins
CA2460621A1 (en) 2001-09-19 2003-03-27 Nuvelo, Inc. Novel nucleic acids and polypeptides
EP2042196B1 (en) 2001-10-10 2016-07-13 ratiopharm GmbH Remodelling and glycoconjugation of Granulocyte Colony Stimulating Factor (G-CSF)
HUP0600342A3 (en) 2001-10-25 2011-03-28 Genentech Inc Glycoprotein compositions
US7897842B2 (en) * 2002-03-19 2011-03-01 Plant Research International B.V. GnTIII expression in plants
IL165717A0 (en) 2002-06-26 2006-01-15 Flanders Interuniversity Inst A strain of methylotrophic yeast for producing proteins
US7514253B2 (en) * 2003-05-16 2009-04-07 Glycofi, Inc. URA5 gene and methods for stable genetic integration in yeast
EP1703803A1 (en) * 2003-12-30 2006-09-27 Gumlink A/S Chewing gum comprising biodegradable polymers and having accelerated degradability
WO2005089047A2 (en) 2004-03-18 2005-09-29 Glycofi, Inc. Glycosylated glucocerebrosidase expression in fungal hosts
US7325756B1 (en) * 2006-09-05 2008-02-05 Giorgis Getachew W Roll-sprinkler irrigation system
US20080274162A1 (en) 2007-05-04 2008-11-06 Jeffrey Nessa Method, composition, and delivery mode for treatment of prostatitis and other urogenital infections using a probiotic rectal suppository

Patent Citations (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4414329A (en) * 1980-01-15 1983-11-08 Phillips Petroleum Company Biochemical conversions by yeast fermentation at high cell densities
US5854018A (en) * 1981-02-25 1998-12-29 Washington Research Foundation Expression of polypeptides in yeast
US4617274A (en) * 1981-10-29 1986-10-14 Phillips Petroleum Company Biochemical conversions by yeast fermentation at high cell densities
US4775622A (en) * 1982-03-08 1988-10-04 Genentech, Inc. Expression, processing and secretion of heterologous protein by yeast
US5955422A (en) * 1983-12-13 1999-09-21 Kirin-Amgen, Inc. Production of erthropoietin
US4879231A (en) * 1984-10-30 1989-11-07 Phillips Petroleum Company Transformation of yeasts of the genus pichia
US4808537A (en) * 1984-10-30 1989-02-28 Phillips Petroleum Company Methanol inducible genes obtained from pichia and methods of use
US4837148A (en) * 1984-10-30 1989-06-06 Phillips Petroleum Company Autonomous replication sequences for yeast strains of the genus pichia
US4855231A (en) * 1984-10-30 1989-08-08 Phillips Petroleum Company Regulatory region for heterologous gene expression in yeast
US4885242A (en) * 1984-10-30 1989-12-05 Phillips Petroleum Company Genes from pichia histidine pathway and uses thereof
US4818700A (en) * 1985-10-25 1989-04-04 Phillips Petroleum Company Pichia pastoris argininosuccinate lyase gene and uses thereof
US4882279A (en) * 1985-10-25 1989-11-21 Phillips Petroleum Company Site selective genomic modification of yeast of the genus pichia
US5032516A (en) * 1985-10-25 1991-07-16 Phillips Petroleum Company Pichia pastoris alcohol oxidase II regulatory region
US5166329A (en) * 1985-10-25 1992-11-24 Phillips Petroleum Company DNA encoding the alcohol oxidase 2 gene of yeast of the genus Pichia
US4935349A (en) * 1986-01-17 1990-06-19 Zymogenetics, Inc. Expression of higher eucaryotic genes in aspergillus
US4812405A (en) * 1986-02-18 1989-03-14 Phillips Petroleum Company Double auxotrophic mutants of Pichia pastoris and methods for preparation
US4925796A (en) * 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US4857467A (en) * 1986-07-23 1989-08-15 Phillips Petroleum Company Carbon and energy source markers for transformation of strains of the genes Pichia
US5002876A (en) * 1986-09-22 1991-03-26 Phillips Petroleum Company Yeast production of human tumor necrosis factor
US4683293A (en) * 1986-10-20 1987-07-28 Phillips Petroleum Company Purification of pichia produced lipophilic proteins
US4929555A (en) * 1987-10-19 1990-05-29 Phillips Petroleum Company Pichia transformation
US5135854A (en) * 1987-10-29 1992-08-04 Zymogenetics, Inc. Methods of regulating protein glycosylation
US5004688A (en) * 1988-04-15 1991-04-02 Phillips Petroleum Company Purification of hepatitis proteins
US5047335A (en) * 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5122465A (en) * 1989-06-12 1992-06-16 Phillips Petroleum Company Strains of pichia pastoris created by interlocus recombination
US5032519A (en) * 1989-10-24 1991-07-16 The Regents Of The Univ. Of California Method for producing secretable glycosyltransferases and other Golgi processing enzymes
US5595900A (en) * 1990-02-14 1997-01-21 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US5324663A (en) * 1990-02-14 1994-06-28 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US5955347A (en) * 1990-02-14 1999-09-21 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US5707828A (en) * 1991-04-25 1998-01-13 Research Corporation Technologies, Inc. Expression of human serum albumin in Pichia pastoris
US5962294A (en) * 1992-03-09 1999-10-05 The Regents Of The University Of California Compositions and methods for the identification and synthesis of sialyltransferases
US5602003A (en) * 1992-06-29 1997-02-11 University Of Georgia Research Foundation N-acetylglucosaminyltransferase V gene
US6096512A (en) * 1993-05-14 2000-08-01 Pharmacia & Upjohn Company Cloned DNA encoding a UDP-GalNAc: Polypeptide, N-acetylgalactosaminyltransferase
US5766910A (en) * 1993-09-09 1998-06-16 La Jolla Cancer Research Foundation Expression of the developmental I antigen by a cloned human cDNA encoding a member of a beta-1, 6-N-acetylglucosaminyltransfrase gene family
US6204431B1 (en) * 1994-03-09 2001-03-20 Abbott Laboratories Transgenic non-human mammals expressing heterologous glycosyltransferase DNA sequences produce oligosaccharides and glycoproteins in their milk
US5861293A (en) * 1994-06-13 1999-01-19 Banyu Pharmaceutical Co., Ltd. Gene encoding glycosyltransferase and its uses
US5945322A (en) * 1994-09-26 1999-08-31 The Rockefeller University Glycosyltransferases for biosynthesis of oligosaccharides, and genes encoding them
US5849904A (en) * 1994-12-22 1998-12-15 Boehringer Mannheim Gmbh Isolated nucleic acid molecules which hybridize to polysialyl transferases
US5834251A (en) * 1994-12-30 1998-11-10 Alko Group Ltd. Methods of modifying carbohydrate moieties
US6017743A (en) * 1995-04-03 2000-01-25 The Institute Of Physical And Chemical Research Siaα 2,3, Galβ 1, 4Glc NAcα 2,8-sialyltransferase
US5910570A (en) * 1995-11-13 1999-06-08 Pharmacia & Upjohn Company Cloned DNA encoding a UDP-GalNAc: polypeptide N-acetylgalactosaminy-ltransferase
US6300113B1 (en) * 1995-11-21 2001-10-09 New England Biolabs Inc. Isolation and composition of novel glycosidases
US5945314A (en) * 1997-03-31 1999-08-31 Abbott Laboratories Process for synthesizing oligosaccharides
US6602684B1 (en) * 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US7214775B2 (en) * 1999-04-09 2007-05-08 Kyowa Hakko Kogyo Co., Ltd. Method of modulating the activity of functional immune molecules
US6410246B1 (en) * 2000-06-23 2002-06-25 Genetastix Corporation Highly diverse library of yeast expression vectors
US7029872B2 (en) * 2000-06-28 2006-04-18 Glycofi, Inc Methods for producing modified glycoproteins
US20060040353A1 (en) * 2000-06-28 2006-02-23 Davidson Robert C Production of galactosylated glycoproteins in lower eukaryotes
US20040230042A1 (en) * 2000-06-28 2004-11-18 Stephen Hamilton Expression of class 2 mannosidase and class III mannosidase in lower eukaryotic cells
US20070105127A1 (en) * 2000-06-28 2007-05-10 Glycofi, Inc. Method for producing modified glycoproteins
US20060286637A1 (en) * 2000-06-28 2006-12-21 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US20060177898A1 (en) * 2000-06-28 2006-08-10 Glycofi, Inc. Methods for producing modified glycoproteins
US20060024304A1 (en) * 2000-06-28 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Man5GlcNAc2 glycoform
US20060029604A1 (en) * 2000-06-28 2006-02-09 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAc2Man3GlcNAc2 glycoform
US20060034828A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAcMAN5GLCNAC2 glycoform
US20060148035A1 (en) * 2000-06-28 2006-07-06 Glycofi, Inc. Methods for producing modified glycoproteins
US20060034830A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GalGlcNAcMan5GLcNAc2 glycoform
US20040018590A1 (en) * 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US20060078963A1 (en) * 2000-06-28 2006-04-13 Glycofi, Inc. Methods for producing modified glycoproteins
US7064191B2 (en) * 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
US20030175884A1 (en) * 2001-08-03 2003-09-18 Pablo Umana Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20060034829A1 (en) * 2001-12-27 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a MAN3GLCNAC2 glycoform
US20060024292A1 (en) * 2001-12-27 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Gal2GlcNAc2Man3GlcNAc2 glycoform
US20050170452A1 (en) * 2001-12-27 2005-08-04 Stefan Wildt Method to engineer mammanlian-type carbohydrate structures
US20040171826A1 (en) * 2003-02-20 2004-09-02 Stephen Hamilton Endomannosidases in the modification of glycoproteins in eukaryotes
US20060160179A1 (en) * 2003-12-24 2006-07-20 Piotr Bobrowicz Methods for eliminating mannosylphosphorylation of glycans in the production of glycoproteins
US20050260729A1 (en) * 2004-03-17 2005-11-24 Hamilton Stephen R Method of engineering a cytidine monophosphate-sialic acid synthetic pathway in fungi and yeast
US20060211085A1 (en) * 2004-04-29 2006-09-21 Piotr Bobrowicz Methods for reducing or eliminating alpha-mannosidase resistant glycans for the production of glycoproteins

Cited By (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080274498A1 (en) * 2000-06-28 2008-11-06 Glycofi, Inc. Methods for producing modified glycoproteins
US8211691B2 (en) 2000-06-28 2012-07-03 Glycofi, Inc. Methods for producing modified glycoproteins
US20060286637A1 (en) * 2000-06-28 2006-12-21 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US20060024304A1 (en) * 2000-06-28 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Man5GlcNAc2 glycoform
US20070105127A1 (en) * 2000-06-28 2007-05-10 Glycofi, Inc. Method for producing modified glycoproteins
US20060029604A1 (en) * 2000-06-28 2006-02-09 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAc2Man3GlcNAc2 glycoform
US7598055B2 (en) 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
US20060034828A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAcMAN5GLCNAC2 glycoform
US7863020B2 (en) 2000-06-28 2011-01-04 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US20060078963A1 (en) * 2000-06-28 2006-04-13 Glycofi, Inc. Methods for producing modified glycoproteins
US20060148035A1 (en) * 2000-06-28 2006-07-06 Glycofi, Inc. Methods for producing modified glycoproteins
US20060177898A1 (en) * 2000-06-28 2006-08-10 Glycofi, Inc. Methods for producing modified glycoproteins
US7629163B2 (en) * 2000-06-28 2009-12-08 Glycofi, Inc. Methods for producing modified glycoproteins
US20040230042A1 (en) * 2000-06-28 2004-11-18 Stephen Hamilton Expression of class 2 mannosidase and class III mannosidase in lower eukaryotic cells
US7935513B2 (en) 2000-06-28 2011-05-03 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US7326681B2 (en) 2000-06-28 2008-02-05 Glycofi, Inc. Methods for producing modified glycoproteins
US20040018590A1 (en) * 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US7923430B2 (en) 2000-06-28 2011-04-12 Glycofi, Inc. Methods for producing modified glycoproteins
US7625756B2 (en) 2000-06-28 2009-12-01 GycoFi, Inc. Expression of class 2 mannosidase and class III mannosidase in lower eukaryotic cells
US7449308B2 (en) 2000-06-28 2008-11-11 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US20090209024A1 (en) * 2000-06-28 2009-08-20 Gerngross Tillman U Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US20070178551A1 (en) * 2000-06-28 2007-08-02 Glycofi, Inc. Methods for producing modified glycoproteins
US8067551B2 (en) 2000-06-28 2011-11-29 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US8932825B2 (en) 2001-12-27 2015-01-13 Glycofi Inc. Method to engineer mammalian-type carbohydrate structures
US20050170452A1 (en) * 2001-12-27 2005-08-04 Stefan Wildt Method to engineer mammanlian-type carbohydrate structures
US20060034829A1 (en) * 2001-12-27 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a MAN3GLCNAC2 glycoform
US20060024292A1 (en) * 2001-12-27 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Gal2GlcNAc2Man3GlcNAc2 glycoform
US20060068035A1 (en) * 2002-03-07 2006-03-30 Pero Ronald W Method of preparation and composition of a water soluble extract of the bioactive component of the plant species Uncaria for enhancing immune, anti-inflammatory, anti-tumor and DNA repair processes of warm blooded animals
US8268609B2 (en) 2003-02-20 2012-09-18 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US8299228B2 (en) 2003-02-20 2012-10-30 Glycofi, Inc. Expression of Class 2 mannosidase and Class III mannosidase in lower eukaryotic cells
US8298811B2 (en) 2003-02-20 2012-10-30 Glycofi, Inc. Expression of Class 2 mannosidase and Class III mannosidase in lower eukaryotic cells
US8999671B2 (en) 2003-02-20 2015-04-07 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US20080085540A1 (en) * 2004-03-17 2008-04-10 Hamilton Stephen R Method of engineering a cytidine monophosphate-sialic acid synthetic pathway in fungi and yeast
US20080199942A1 (en) * 2004-03-17 2008-08-21 Hamilton Stephen R Method of engineering a cytidine monophosphate-sialic acid synthetic pathway in fungi and yeast
US20090136525A1 (en) * 2005-09-02 2009-05-28 Tillman Gerngross Immunoglobulins Comprising Predominantly a Glcnacman3Glcnac2 Glycoform
US20090226464A1 (en) * 2005-09-09 2009-09-10 Tillman Gerngross Immunoglobulins comprising predominantly a glcnacman3glcnac2 glycoform
US8685671B2 (en) 2005-10-27 2014-04-01 Korea Research Institute Of Bioscience And Biotechnology Process for producing recombinant glycoproteins by culturing a Hansenula polymorpha mutant strain
US8187858B2 (en) * 2005-10-27 2012-05-29 Korea Research Institute Of Bioscience And Biotechnology Hansenula polymorpha gene coding for dolichyl-phosphate-mannose dependent alpha-1,3-mannosyltransferase and process for the production of recombinant glycoproteins with Hansenula polymorpha mutant strain deficient in the same gene
US20100062485A1 (en) * 2005-10-27 2010-03-11 Korea Research Institute Of Bioscience And Biotechnology Novel Hansenula Polymorpha Gene Coding for Dolichyl-Phosphate- Mannose Dependent Alpha-1,3 Mannosyltransferase and Process for the Production of Recombinant Glycoproteins With Hansenula Polymorpha Mutant Strain Deficient in
US20090170159A1 (en) * 2005-11-15 2009-07-02 Glycofi, Inc Production of glycoproteins with reduced o-glycosylation
US8206949B2 (en) 2005-11-15 2012-06-26 Glycofi, Inc. Production of glycoproteins with reduced O-glycosylation
US8795984B2 (en) 2005-11-15 2014-08-05 Merck Sharp & Dohme Corp. Production of glycoproteins with reduced O-glycosylation
US8501438B2 (en) 2005-11-15 2013-08-06 Glycofi, Inc. Production of glycoproteins with reduced O-glycosylation
AU2007248485B2 (en) * 2006-05-05 2013-01-24 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
EP2027281A2 (en) * 2006-05-05 2009-02-25 Glycofi, Inc. Production of sialylated n-glycans in lower eukaryotes
EP2027281A4 (en) * 2006-05-05 2010-03-10 Glycofi Inc Production of sialylated n-glycans in lower eukaryotes
JP2009536031A (en) * 2006-05-05 2009-10-08 グライコフィ, インコーポレイテッド Production of sialylated N-glycans in lower eukaryotes
US8741633B2 (en) 2006-05-19 2014-06-03 Glycofi, Inc. Recombinant vectors
US20100028951A1 (en) * 2007-03-07 2010-02-04 Stephen Hamilton Production of glycoproteins with modified fucosylation
EP2546347A2 (en) 2009-02-25 2013-01-16 Merck Sharp & Dohme Corp. Glycoprotein composition from engineered galactose assimilation pathway in Pichia pastoris
US20120283420A1 (en) * 2009-04-22 2012-11-08 Bieke Nagels Production of Multi-Antennary N-Glycan Structures in Plants
US8715963B2 (en) 2010-02-24 2014-05-06 Merck Sharp & Dohme Corp. Method for increasing N-glycosylation site occupancy on therapeutic glycoproteins produced in Pichia pastoris
WO2011106389A1 (en) 2010-02-24 2011-09-01 Merck Sharp & Dohme Corp. Method for increasing n-glycosylation site occupancy on therapeutic glycoproteins produced in pichia pastoris
US10858686B2 (en) 2010-05-27 2020-12-08 Merck Sharp & Dohme Corp. Method for preparing antibodies having improved properties
US9328170B2 (en) 2011-05-25 2016-05-03 Merck Sharp & Dohme Corp. Method for preparing Fc containing polypeptides having improved properties
US9695454B2 (en) 2012-05-23 2017-07-04 Glykos Finland Oy Production of fucosylated glycoproteins
WO2014064203A1 (en) 2012-10-26 2014-05-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Lyve-1 antagonists for preventing or treating a pathological condition associated with lymphangiogenesis
WO2014090948A1 (en) 2012-12-13 2014-06-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Serpin spn4a and biologically active derivatives thereof for use in the treatment of cancer
WO2014111744A1 (en) 2013-01-16 2014-07-24 INSERM (Institut National de la Santé et de la Recherche Médicale) A soluble fibroblast growth factor receptor 3 (fgr3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
WO2014111467A1 (en) 2013-01-16 2014-07-24 INSERM (Institut National de la Santé et de la Recherche Médicale) A soluble fibroblast growth factor receptor 3 (fgr3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
EP3736285A1 (en) 2013-01-16 2020-11-11 INSERM (Institut National de la Santé et de la Recherche Médicale) A soluble fibroblast growth factor receptor 3 (fgr3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
WO2015044379A1 (en) 2013-09-27 2015-04-02 INSERM (Institut National de la Santé et de la Recherche Médicale) A dyrk1a polypeptide for use in preventing or treating metabolic disorders
US10513724B2 (en) 2014-07-21 2019-12-24 Glykos Finland Oy Production of glycoproteins with mammalian-like N-glycans in filamentous fungi
CN112041456A (en) * 2018-05-01 2020-12-04 科纳根公司 Recombinant organisms and methods for producing sugar molecules with low sulfation
US11959118B2 (en) 2020-10-20 2024-04-16 Merck Sharp & Dohme Llc Fc-containing polypeptides having improved properties and comprising mutations at positions 243 and 264 of the Fc-region
EP4309666A1 (en) 2022-07-21 2024-01-24 Technische Universität Dresden M-csf for use in the prophylaxis and/or treatment of viral infections in states of immunosuppression
WO2024017998A1 (en) 2022-07-21 2024-01-25 Technische Universität Dresden M-csf for use in the prophylaxis and/or treatment of viral infections in states of immunosuppression

Also Published As

Publication number Publication date
EP1599595B1 (en) 2009-11-25
SI1599595T1 (en) 2010-03-31
JP2011019521A (en) 2011-02-03
AU2010201036A1 (en) 2010-04-08
EP1599595B2 (en) 2013-08-21
AU2004213868A1 (en) 2004-09-02
WO2004074461A2 (en) 2004-09-02
AU2004213859B2 (en) 2010-01-07
US8445227B2 (en) 2013-05-21
PT1597381E (en) 2010-02-23
CA2516520A1 (en) 2004-09-02
DE602004024289D1 (en) 2010-01-07
ES2336790T5 (en) 2013-09-05
ES2335345T5 (en) 2013-12-04
CA2516520C (en) 2013-10-01
ES2335345T3 (en) 2010-03-25
ES2336790T3 (en) 2010-04-16
WO2004074458A3 (en) 2004-12-29
WO2004074458A2 (en) 2004-09-02
SI1597381T2 (en) 2013-08-30
PT1599595E (en) 2010-01-26
ATE543912T1 (en) 2012-02-15
JP4611280B2 (en) 2011-01-12
ATE452201T1 (en) 2010-01-15
EP2080809B1 (en) 2012-02-01
JP2006518600A (en) 2006-08-17
AU2004213868B2 (en) 2010-05-20
WO2004074461A3 (en) 2005-03-17
ATE449862T1 (en) 2009-12-15
US20100016561A1 (en) 2010-01-21
AU2010201036B2 (en) 2011-12-01
JP4758335B2 (en) 2011-08-24
JP2006518597A (en) 2006-08-17
CA2516550C (en) 2013-12-24
CA2753408A1 (en) 2004-09-02
EP1597381B1 (en) 2009-12-16
CA2516550A1 (en) 2004-09-02
DE602004024649D1 (en) 2010-01-28
EP2080809A1 (en) 2009-07-22
SI1599595T2 (en) 2013-12-31
EP1597381B2 (en) 2013-05-15
EP1599595A2 (en) 2005-11-30
EP1597381A2 (en) 2005-11-23
US7598055B2 (en) 2009-10-06
JP5255017B2 (en) 2013-08-07
SI1597381T1 (en) 2010-04-30
ES2384929T3 (en) 2012-07-16
JP2010207236A (en) 2010-09-24
AU2004213859A1 (en) 2004-09-02
US20100016555A1 (en) 2010-01-21

Similar Documents

Publication Publication Date Title
US7598055B2 (en) N-acetylglucosaminyltransferase III expression in lower eukaryotes
US8697394B2 (en) Production of modified glycoproteins having multiple antennary structures
US8067551B2 (en) Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US8299228B2 (en) Expression of Class 2 mannosidase and Class III mannosidase in lower eukaryotic cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLYCOFI, INC., NEW HAMPSHIRE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BOBROWICZ, PIOTR;HAMILTON, STEPHEN R.;GERNGROSS, TILLMAN U.;AND OTHERS;REEL/FRAME:018595/0636;SIGNING DATES FROM 20061113 TO 20061206

FPAY Fee payment

Year of fee payment: 4

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.)

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20171006